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:

Structure of SWI/SNF chromatin remodeller RSC bound to a nucleosome

Abstract

Chromatin-remodelling complexes of the SWI/SNF family function in the formation of nucleosome-depleted, transcriptionally active promoter regions (NDRs)1,2. In the yeast Saccharomyces cerevisiae, the essential SWI/SNF complex RSC3 contains 16 subunits, including the ATP-dependent DNA translocase Sth14,5. RSC removes nucleosomes from promoter regions6,7 and positions the specialized +1 and −1 nucleosomes that flank NDRs8,9. Here we present the cryo-electron microscopy structure of RSC in complex with a nucleosome substrate. The structure reveals that RSC forms five protein modules and suggests key features of the remodelling mechanism. The body module serves as a scaffold for the four flexible modules that we call DNA-interacting, ATPase, arm and actin-related protein (ARP) modules. The DNA-interacting module binds extra-nucleosomal DNA and is involved in the recognition of promoter DNA elements8,10,11 that influence RSC functionality12. The ATPase and arm modules sandwich the nucleosome disc with the Snf2 ATP-coupling (SnAC) domain and the finger helix, respectively. The translocase motor of the ATPase module engages with the edge of the nucleosome at superhelical location +2. The mobile ARP module may modulate translocase–nucleosome interactions to regulate RSC activity5. The RSC–nucleosome structure provides a basis for understanding NDR formation and the structure and function of human SWI/SNF complexes that are frequently mutated in cancer13.

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: RSC–nucleosome complex structure.
Fig. 2: RSC ATPase and ARP modules.
Fig. 3: RSC sandwiches the nucleosome.

Similar content being viewed by others

Data availability

The coordinate file for the RSC–nucleosome complex structure was deposited with the Protein Data Bank with accession code 6TDA. The cryo-EM maps used for model building were all deposited with the Electron Microscopy Data Base with accession code EMD-10465.

References

  1. Lorch, Y. & Kornberg, R. D. Chromatin-remodeling for transcription. Q. Rev. Biophys. 50, e5 (2017).

    PubMed  Google Scholar 

  2. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Cairns, B. R. et al. RSC, an essential, abundant chromatin-remodeling complex. Cell 87, 1249–1260 (1996).

    CAS  PubMed  Google Scholar 

  4. Saha, A., Wittmeyer, J. & Cairns, B. R. Chromatin remodeling by RSC involves ATP-dependent DNA translocation. Genes Dev. 16, 2120–2134 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Clapier, C. R. et al. Regulation of DNA translocation efficiency within the chromatin remodeler RSC/Sth1 potentiates nucleosome sliding and ejection. Mol. Cell 62, 453–461 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Krietenstein, N. et al. Genomic nucleosome organization reconstituted with pure proteins. Cell 167, 709–712 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Klein-Brill, A., Joseph-Strauss, D., Appleboim, A. & Friedman, N. Dynamics of chromatin and transcription during transient depletion of the RSC chromatin remodeling complex. Cell Rep. 26, 279–292 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Kubik, S. et al. Nucleosome stability distinguishes two different promoter types at all protein-coding genes in yeast. Mol. Cell 60, 422–434 (2015).

    CAS  PubMed  Google Scholar 

  9. Ramachandran, S., Zentner, G. E. & Henikoff, S. Asymmetric nucleosomes flank promoters in the budding yeast genome. Genome Res. 25, 381–390 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Badis, G. et al. A library of yeast transcription factor motifs reveals a widespread function for Rsc3 in targeting nucleosome exclusion at promoters. Mol. Cell 32, 878–887 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Lorch, Y., Maier-Davis, B. & Kornberg, R. D. Role of DNA sequence in chromatin remodeling and the formation of nucleosome-free regions. Genes Dev. 28, 2492–2497 (2014).

    PubMed  PubMed Central  Google Scholar 

  12. Kubik, S. et al. Sequence-directed action of RSC remodeler and general regulatory factors modulates +1 nucleosome position to facilitate transcription. Mol Cell 71, 89–102 (2018).

    CAS  PubMed  Google Scholar 

  13. Pulice, J. L. & Kadoch, C. Composition and function of mammalian SWI/SNF chromatin remodeling complexes in human disease. Cold Spring Harb. Symp. Quant. Biol. 81, 53–60 (2016).

    PubMed  Google Scholar 

  14. Chambers, A. L., Pearl, L. H., Oliver, A. W. & Downs, J. A. The BAH domain of Rsc2 is a histone H3 binding domain. Nucleic Acids Res. 41, 9168–9182 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kasten, M. et al. Tandem bromodomains in the chromatin remodeler RSC recognize acetylated histone H3 Lys14. EMBO J. 23, 1348–1359 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. VanDemark, A. P. et al. Autoregulation of the Rsc4 tandem bromodomain by Gcn5 acetylation. Mol. Cell 27, 817–828 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Li, M. et al. Mechanism of DNA translocation underlying chromatin remodelling by Snf2. Nature 567, 409–413 (2019).

    ADS  CAS  PubMed  Google Scholar 

  18. Saha, A., Wittmeyer, J. & Cairns, B. R. Chromatin remodeling through directional DNA translocation from an internal nucleosomal site. Nat. Struct. Mol. Biol. 12, 747–755 (2005).

    CAS  PubMed  Google Scholar 

  19. Schubert, H. L. et al. Structure of an actin-related subcomplex of the SWI/SNF chromatin remodeler. Proc. Natl Acad. Sci. USA 110, 3345–3350 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Szerlong, H. et al. The HSA domain binds nuclear actin-related proteins to regulate chromatin-remodeling ATPases. Nat. Struct. Mol. Biol. 15, 469–476 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Sen, P. et al. The SnAC domain of SWI/SNF is a histone anchor required for remodeling. Mol. Cell. Biol. 33, 360–370 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lorch, Y., Maier-Davis, B. & Kornberg, R. D. Histone acetylation inhibits RSC and stabilizes the +1 nucleosome. Mol. Cell. 72, 594–600 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Cakiroglu, A. et al. Genome-wide reconstitution of chromatin transactions reveals that RSC preferentially disrupts H2AZ-containing nucleosomes. Genome Res. 29, 988–998 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Suto, R. K., Clarkson, M. J., Tremethick, D. J. & Luger, K. Crystal structure of a nucleosome core particle containing the variant histone H2A.Z. Nat. Struct. Biol. 7, 1121–1124 (2000).

    CAS  PubMed  Google Scholar 

  25. Materne, P. et al. Histone H2B ubiquitylation represses gametogenesis by opposing RSC-dependent chromatin remodeling at the ste11 master regulator locus. eLife 5, e13500 (2016).

  26. Brahma, S. & Henikoff, S. RSC-associated subnucleosomes define MNase-sensitive promoters in yeast. Mol. Cell 73, 238–249 (2019).

    CAS  PubMed  Google Scholar 

  27. Brogaard, K., Xi, L., Wang, J. P. & Widom, J. A map of nucleosome positions in yeast at base-pair resolution. Nature 486, 496–501 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dechassa, M. L. et al. SWI/SNF has intrinsic nucleosome disassembly activity that is dependent on adjacent nucleosomes. Mol. Cell 38, 590–602 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Patel, A. B. et al. Architecture of the chromatin remodeler RSC and insights into its nucleosome engagement. eLife 8, e54449 (2019).

    PubMed  PubMed Central  Google Scholar 

  30. Ye, Y. et al. Structure of the RSC complex bound to the nucleosome. Science 366, 838–843 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Han, Y., Reyes, A. A., Malik, S. & He, Y. Cryo-electron microscopy structure of a nucleosome-bound SWI/SNF chromatin remodeling complex. Preprint at https://www.bioRxiv.org/content/10.1101/805184v1 (2019).

  32. Cairns, B. R. et al. Two functionally distinct forms of the RSC nucleosome-remodeling complex, containing essential AT hook, BAH, and bromodomains. Mol. Cell 4, 715–723 (1999).

    CAS  PubMed  Google Scholar 

  33. Rigaut, G. et al. A generic protein purification method for protein complex characterization and proteome exploration. Nat. Biotechnol. 17, 1030–1032 (1999).

    CAS  PubMed  Google Scholar 

  34. Lorch, Y. & Kornberg, R. D. Isolation and assay of the RSC chromatin-remodeling complex from Saccharomyces cerevisiae. Methods Enzymol. 377, 316–322 (2003).

    Google Scholar 

  35. Luger, K., Rechsteiner, T. J. & Richmond, T. J. Expression and purification of recombinant histones and nucleosome reconstitution. Methods Mol. Biol. 119, 1–16 (1999).

    CAS  PubMed  Google Scholar 

  36. Dyer, P. N. et al. Reconstitution of nucleosome core particles from recombinant histones and DNA. Methods Enzymol. 375, 23–44 (2003).

    Google Scholar 

  37. Maskell, D. P. et al. Structural basis for retroviral integration into nucleosomes. Nature 523, 366–369 (2015).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  40. Stark, H. GraFix: stabilization of fragile macromolecular complexes for single particle cryo-EM. Methods Enzymol. 481, 109–126 (2010).

    CAS  PubMed  Google Scholar 

  41. Tegunov, D. & Cramer, P. Real-time cryo-EM data pre-processing with Warp. Nat. Methods 16, 1146–1152 (2019).

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  44. Asturias, F. J., Chung, W. H., Kornberg, R. D. & Lorch, Y. Structural analysis of the RSC chromatin-remodeling complex. Proc. Natl Acad. Sci. USA 99, 13477–13480 (2002).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chaban, Y. et al. Structure of a RSC-nucleosome complex and insights into chromatin remodeling. Nat. Struct. Mol. Biol. 15, 1272–1277 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Leschziner, A. E. et al. Conformational flexibility in the chromatin remodeler RSC observed by electron microscopy and the orthogonal tilt reconstruction method. Proc. Natl Acad. Sci. USA 104, 4913–4918 (2007).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Kidmose, R. T. et al. Namdinator—automatic molecular dynamics flexible fitting of structural models into cryo-EM and crystallography experimental maps. IUCrJ 6, 526–531 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Song, Y. et al. High-resolution comparative modeling with RosettaCM. Structure 21, 1735–1742 (2013).

    CAS  PubMed  Google Scholar 

  51. Raman, S. et al. Structure prediction for CASP8 with all-atom refinement using Rosetta. Proteins 77 (Suppl 9), 89–99 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. van Dijk, M. & Bonvin, A. M. 3D-DART: a DNA structure modelling server. Nucleic Acids Res. 37, W235–W239 (2009).

    PubMed  PubMed Central  Google Scholar 

  53. Waterhouse, A. et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 46, W296–W303 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Bienert, S. et al. The SWISS-MODEL repository-new features and functionality. Nucleic Acids Res. 45, D313–D319 (2017).

    CAS  PubMed  Google Scholar 

  55. Charlop-Powers, Z., Zeng, L., Zhang, Q. & Zhou, M. M. Structural insights into selective histone H3 recognition by the human Polybromo bromodomain 2. Cell Res. 20, 529–538 (2010).

    CAS  PubMed  Google Scholar 

  56. Da, G. et al. Structure and function of the SWIRM domain, a conserved protein module found in chromatin regulatory complexes. Proc. Natl Acad. Sci. USA 103, 2057–2062 (2006).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  57. Legge, G. B. et al. ZZ domain of CBP: an unusual zinc finger fold in a protein interaction module. J. Mol. Biol. 343, 1081–1093 (2004).

    CAS  PubMed  Google Scholar 

  58. Reichen, C. et al. Structures of designed armadillo-repeat proteins show propagation of inter-repeat interface effects. Acta Crystallogr. D 72, 168–175 (2016).

    CAS  Google Scholar 

  59. Grimm, M., Zimniak, T., Kahraman, A. & Herzog, F. xVis: a web server for the schematic visualization and interpretation of crosslink-derived spatial restraints. Nucleic Acids Res. 43, W362–W369 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Zimmermann, L. et al. A completely reimplemented mpi bioinformatics toolkit with a new HHpred server at its core. J. Mol. Biol. 430, 2237–2243 (2018).

    CAS  PubMed  Google Scholar 

  61. Buchan, D. W. A. & Jones, D. T. The PSIPRED protein analysis workbench: 20 years on. Nucleic Acids Res. 47, W402–W407 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Jones, D. T. Protein secondary structure prediction based on position-specific scoring matrices. J. Mol. Biol. 292, 195–202 (1999).

    CAS  PubMed  Google Scholar 

  63. Williams, C. J. et al. MolProbity: More and better reference data for improved all-atom structure validation. Protein Sci. 27, 293–315 (2018).

    CAS  PubMed  Google Scholar 

  64. Schrodinger, LLC. The PyMOL Molecular Graphics System version 1.8 (2015).

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

    CAS  PubMed  Google Scholar 

  66. Treich, I., Ho, L. & Carlson, M. Direct interaction between Rsc6 and Rsc8/Swh3,two proteins that are conserved in SWI/SNF-related complexes. Nucleic Acids Res. 26, 3739–3745 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Taneda, T. & Kikuchi, A. Genetic analysis of RSC58, which encodes a component of a yeast chromatin remodeling complex, and interacts with the transcription factor Swi6. Mol. Genet. Genomics 271, 479–489 (2004).

    CAS  PubMed  Google Scholar 

  68. Angus-Hill, M. L. et al. A Rsc3/Rsc30 zinc cluster dimer reveals novel roles for the chromatin remodeler RSC in gene expression and cell cycle control. Mol. Cell 7, 741–751 (2001).

    CAS  PubMed  Google Scholar 

  69. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    PubMed  Google Scholar 

  70. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  71. Liu, Y., Schmidt, B. & Maskell, D. L. MSAProbs: multiple sequence alignment based on pair hidden Markov models and partition function posterior probabilities. Bioinformatics 26, 1958–1964 (2010).

    CAS  PubMed  Google Scholar 

  72. Bond, C. S. & Schüttelkopf, A. W. ALINE: a WYSIWYG protein-sequence alignment editor for publication-quality alignments. Acta Crystallogr. D 65, 510–512 (2009).

    CAS  PubMed  Google Scholar 

  73. Yang, B. et al. Identification of cross-linked peptides from complex samples. Nat. Methods 9, 904–906 (2012).

    CAS  PubMed  Google Scholar 

  74. Combe, C. W., Fischer, L. & Rappsilber, J. xiNET: cross-link network maps with residue resolution. Mol. Cell. Proteomics 14, 1137–1147 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Yan, Z. et al. PBAF chromatin-remodeling complex requires a novel specificity subunit, BAF200, to regulate expression of selective interferon-responsive genes. Genes Dev. 19, 1662–1667 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Xue, Y. et al. The human SWI/SNF-B chromatin-remodeling complex is related to yeast rsc and localizes at kinetochores of mitotic chromosomes. Proc. Natl Acad. Sci. USA 97, 13015–13020 (2000).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  77. Nicolas, R. H. & Goodwin, G. H. Molecular cloning of polybromo, a nuclear protein containing multiple domains including five bromodomains, a truncated HMG-box, and two repeats of a novel domain. Gene 175, 233–240 (1996).

    CAS  PubMed  Google Scholar 

  78. Wang, W. et al. Architectural DNA binding by a high-mobility-group/kinesin-like subunit in mammalian SWI/SNF-related complexes. Proc. Natl Acad. Sci. USA 95, 492–498 (1998).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  79. Satterwhite, E. et al. The BCL11 gene family: involvement of BCL11A in lymphoid malignancies. Blood 98, 3413–3420 (2001).

    CAS  PubMed  Google Scholar 

  80. Sandhya, S., Maulik, A., Giri, M. & Singh, M. Domain architecture of BAF250a reveals the ARID and ARM-repeat domains with implication in function and assembly of the BAF remodeling complex. PLoS One 13, e0205267 (2018).

    PubMed  PubMed Central  Google Scholar 

  81. 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 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Eustermann, S. et al. Structural basis for ATP-dependent chromatin remodelling by the INO80 complex. Nature 556, 386–390 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  83. Farnung, L., Vos, S. M., Wigge, C. & Cramer, P. Nucleosome–Chd1 structure and implications for chromatin remodelling. Nature 550, 539–542 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  84. Willhoft, O. et al. Structure and dynamics of the yeast SWR1-nucleosome complex. Science 362, eaat7716 (2018).

    PubMed  Google Scholar 

  85. Ayala, R. et al. Structure and regulation of the human INO80-nucleosome complex. Nature 556, 391–395 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  86. Sundaramoorthy, R. et al. Structure of the chromatin remodelling enzyme Chd1 bound to a ubiquitinylated nucleosome. eLife 7, e35720 (2018).

    PubMed  PubMed Central  Google Scholar 

  87. Yan, L., Wu, H., Li, X., Gao, N. & Chen, Z. Structures of the ISWI-nucleosome complex reveal a conserved mechanism of chromatin remodeling. Nat. Struct. Mol. Biol. 26, 258–266 (2019).

    CAS  PubMed  Google Scholar 

  88. Robert, X. & Gouet, P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res. 42, W320–W324 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank current and former members of the Cramer Laboratory, including S. Osman, G. Kokic, P. Seweryn, S. Schilbach, S. Neyer and H. Hillen. F.R.W. was supported by a Boehringer Ingelheim Fonds PhD fellowship. H.U. was supported by the Deutsche Forschungsgemeinschaft (SFB860). P.C. was supported by the Deutsche Forschungsgemeinschaft (SFB860, SPP1935, EXC 2067/1-390729940), the European Research Council Advanced Investigator Grant TRANSREGULON (grant agreement no. 693023), and the Volkswagen Foundation.

Author information

Authors and Affiliations

Authors

Contributions

F.R.W. carried out all experiments and data analysis unless stated otherwise. C.D. assisted with data collection and model building. A.S. and H.U. carried out cross-linking and mass spectrometry analysis. H.W. helped with nucleosome biochemistry. D.T. helped with cryo-EM data processing. P.C. designed and supervised the project. F.W. and P.C. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Patrick Cramer.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Blaine Bartholomew and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Preparation and characterization of RSC–nucleosome complex.

a, Preparation of endogenous Rsc2-containing isoform of the RSC complex from S. cerevisiae. Analysis of purified RSC by size-exclusion chromatography and SDS–PAGE showed high purity and homogeneity with stoichiometric subunits as assessable by Coomassie stain. Subunit identity was confirmed by mass spectrometry. The table shows the expected molecular weights of the RSC subunits. For gel source data, see Supplementary Fig. 1. b, Assembly of the RSC–nucleosome complex. SDS–PAGE analysis of fractions 7–20 of a 10–25% sucrose-gradient ultracentrifugation. Complex formation was successful as demonstrated by the comigration of histones with the RSC complex. The unbound over-stoichiometric nucleosomes only migrated to fractions 7 and 8 (black arrow). Fraction 16 in the presence of cross-linker was used for cryo-EM grid preparation (dashed box). c, Location of cross-linking sites mapped onto the structure. BS3 cross-links that appeared at least in triplicates were mapped onto the RSC-nucleosome structure. Lysine residues involved in the cross-linking network are shown as blue spheres and cross-linked residues are connected with lines indicating permitted (blue) and non-permitted (red) cross-linking distances. Of the mapped cross-links, 87.5% are within the permitted cross-linking distance, which was set to 30 Å. The remaining 12.5% of non-permitted cross-links probably reflect ambiguity caused by the presence of two identical Rsc8 subunits in the structure as well as flexibility of the complex in buffer or arise from technical errors. d, The cross-linking network between subunits of the RSC–nucleosome complex. Subunits are coloured as in Fig. 1. Cross-links with a score above 2.5 are shown. A comprehensive list of cross-links is presented in Supplementary Data 1. Cross-linking mass-spectrometry experiments were performed in duplicates with similar results.

Extended Data Fig. 2 Cryo-EM analysis of the RSC–nucleosome complex.

a, Representative cryo-EM micrograph of the RSC–nucleosome complex shows homogeneously distributed individual particles. bd, Two-dimensional class averages of the RSC–nucleosome complex (b), the ATPase–nucleosome subcomplex (c) and the nucleosome subcomplex (d). e, FSC plots reveal the overall resolutions of the cryo-EM reconstructions. f, Cryo-EM processing workflow for the reconstructions of the RSC–nucleosome complex, the ATPase–nucleosome subcomplex and the nucleosome subcomplex. Particle distribution after 3D classifications is indicated below the corresponding map. The final maps are shown in colour. The masks used for focused classifications and refinements are colour coded corresponding to the final maps they were used for. Views are generally rotated by 180° with respect to Fig. 1c, left. g, Local resolution estimation of the combined ATPase–nucleosome map as implemented in RELION42. We note that the resolution of the peripheral area with the ATPase module is overestimated. hj, Angular distribution plot for all particles contributing to the final reconstructions of the RSC–nucleosome complex (h), the ATPase–nucleosome subcomplex (i) and the nucleosome subcomplex (j).

Extended Data Fig. 3 Cryo-EM analysis of the free RSC complex.

a, Representative cryo-EM micrograph of the free RSC complex shows homogeneously distributed individual particles. b, Two-dimensional class averages of the free RSC complex. c, Cryo-EM processing workflow for the reconstruction of the free RSC complex. Particle distribution after 3D classifications is indicated below the corresponding map. The final maps after focused 3D refinement and masks are depicted in colour. Views are generally rotated by 180° with respect to Fig. 1c, right. d, Angular distribution plot for all particles contributing to the final reconstruction of the free RSC complex. e, Two views of the combined RSC core map coloured according to the local resolution as implemented in RELION42. f, FSC plots of the maps used for model building of the RSC core complex.

Extended Data Fig. 4 Cryo-EM densities for selected RSC regions.

ac, Examples of map quality. a, Close-up of the Rsc4 β-sheet shows clear separation of individual strands. b, The high quality of the map for the ZZ zinc-finger of Rsc8 allowed backbone tracing and placement of side chains as well as for the zinc ion. c, Coiled-coil helices of the two Rsc8 subunits with density for one helix. d, View along the exit DNA in the direction of the nucleosome showing the lowpass-filtered maps for the modules ATPase, ARP, DIM, arm and body, and the nucleosome. At the site where the H2A C-terminal tail protrudes from the nucleosome near Sfh1, there is low-resolution density connecting the arm module and the nucleosome. Density bridging form the ARP module to the exit DNA close to the H3 histone tail can be observed. e, Density representing the finger helix (green) at the acidic patch of the nucleosome (indicated by H2A in yellow). Side-chain density is visible for conserved arginine residues. f, Interaction of RSC with the nucleosome is sterically impaired by the flexibly bound ubiquitin moiety at H2B lysine 123 (K120 in human). The Sfh1 finger helix and the ubiquitin moiety (ubiquitylated nucleosome PDB code 6NOG)81 overlap after superposition of nucleosomes.

Extended Data Fig. 5 Structure of RSC body and arm modules, cancer mutations and remodeller families.

a, Cartoon representation of RSC core viewed as in Fig. 1. Important structural elements are labelled. b, Conservation between SWI/SNF complexes RSC (yeast) and PBAF (human). Residues that are identical (blue) or conserved (light blue) in human PBAF highlighted on the RSC structure (grey). Purple spheres depict identical residues that show missense mutations in various cancers. c, Comparison of overall structure of RSC with complexes of INO80 (yeast INO80)82 and CHD (yeast CHD1)83 families. ATPase motor domains are shown in orange, DNA is shown in blue. With regard to the INO80 family, the ATPase of the SWR1 complex also binds SHL +2 (ref. 84), whereas the ATPase of the INO80 complex binds SHL −6 (refs. 82,85). The ARP module of INO80 contacts exit DNA, which is not the case in RSC. The INO80 complex also contacts both faces of the histone octamer82, resembling the sandwiching interactions made by RSC on a topological level. With respect to the CHD family, the ATPase motor of yeast Chd1 also binds SHL +2, but its DNA-binding region engages with exit DNA near the nucleosome, leading to a different DNA trajectory83,86. With respect to the ISWI family, the ATPase motor binds SHL +2 (ref. 87), but other interactions have not been structurally resolved (not shown).

Extended Data Fig. 6 Course of polypeptide chains of architectural subunits Sth1, Rsc8 and Rsc58 and ATPase–nucleosome interactions.

a, Back view of RSC. The Sth1 subunit of RSC starts with its N terminus in the body module and tracks through it, turning around with a contact helix and loop. Forming the central helix I, the hook and the central helix II it folds back and forth tightly interweaving the body module before it exits with its HSA region through the ARP module to build the ATPase module. b, RSC with the domains of the two Rsc8 subunits highlighted in blue. Both Rsc8 start N-terminal with their SWIRM domains in the arm module where they support the two repeat domains of Sfh1 in a similar manner. They then follow distinct paths through the arm towards the body module where they contribute with both their SANT and ZZ zinc-finger domains. Here the two domains of each subunit form different contacts with various interactions partners and whereas one ZZ zinc-finger domain is tightly packed at the body and DNA-interaction module interface, the other seems to extend from the body, presumably as additional interaction surface. Both Rsc8 subunits unite again with their C-terminal long helices in a coiled-coil fold on the opposite side of the body module. c, Rsc58 N-terminal bromodomain attaches to the top of the body module. Then, Rsc58 follows an interwound path through the body module via the central and connector loop. It turns back, docking to the body with a three-helix bundle and stabilizing the module with its C-terminal end. d, Contacts of Sth1 ATPase motor (orange) with the nucleosome. View as in Fig. 1c, left, but rotated by 45° around a horizontal axis. Arrows indicate directionality of DNA translocation.

Extended Data Fig. 7 DNA recognition and NDR formation.

a, Space-filling RSC–nucleosome structure with DIM (green) and SnAC (orange) densities. View on the top as in Fig. 1c, left, but rotated by 90° around the vertical and horizontal axis. Arrows indicate directionality of DNA translocation. Number of upstream DNA base pairs relative to SHL −7 is provided. b, Schematic of a promoter before (top) and after (bottom) RSC remodelling shows NDR formation by sliding the flanking –1 and +1 nucleosomes away from the NDR centre. Arrows indicate the transcription start site.

Extended Data Fig. 8 Sequence alignments for the Sth1 ATPase domain and HSA region.

a, Sequence alignment of the S. cerevisiae Sth1 ATPase domain to the homologous Snf2 ATPase domain of the same organism. Secondary structure elements are represented in orange according to the cryo-EM structure of the Snf2 ATPase (PDB entry 5Z3U)17. Residues modelled in the Snf2 structure are topped by a black line with helical regions shown as cylinders and sheet regions as arrows. The Sth1 residues modelled in this work are indicated with a black dashed line below. ATPase motifs are underlined. Invariant residues are coloured in dark blue and conserved residues in light blue. The alignment was generated with MSAProbs71 within the MPI Bioinformatics Toolkit60 and visualized using ESPript88. b, Sequence alignment of the HSA regions from S. cerevisiae homologues Sth1 and Snf2. Illustration and generation of the alignment as in a.

Extended Data Table 1 Subunit composition of RSC and related chromatin remodelling complexes

Supplementary information

Supplementary Figure

Supplementary Figure 1 | Uncropped gel images with size marker. Uncropped gel images for Extended Data Figs. 1a and 1b. * Sample from another unrelated experiment, not from the size exclusion chromatography run.

Reporting Summary

Supplementary Table

Supplementary Table 1 | RSC subunit modelling. Modelling details for the RSC complex. Density that could not be assigned to a subunit was modelled with a poly alanine backbone in chain X. The domains of the two Rsc8 subunits could not be connected. Therefore, they are clustered by proximity and combined in one chain (L) spaced by a 443 amino acid number offset.

Supplementary Table

Supplementary Table 2 | Cryo-EM data collection, refinement and validation statistics. * Dataset from two collections were combined. Datasets from three collections were combined, not tilted, 25° tilt. # Reported value is based on visual inspection, because the software-generated values were overestimated. § Map7: chains A, B, C, D, E, F, G, H, I (2-124), J (24-146), K (390-408) and S (393-1006); Map8: chains K (151-389), L, M, N, O, P, Q, R, S (48-297) and X.

Supplementary Data

Supplementary Data 1 | RSC-nucleosome BS3 crosslinks. List of intra- and inter-subunit lysine-lysine crosslinks as identified by LC-MS analyses and subsequent database search using pLink 2. The respective scores of cross-link identification are listed as well as the number of CSMs (cross-linked spectra matches). The numbering for the histone H3 is shifted because the N-terminal PEP sequence was omitted for purification (instead of MPEPAKSAP... it is MAKSAP...). The numbering for the Rsc2 protein includes the remains of C-terminal TAP-tag after TEV cleavage.

Video 1

Overview of RSC structure. The video shows the structure of RSC rotating around a vertical axis. It first depicts the low pass-filtered cryo-EM  map, showing the five lobes of RSC and the nucleosome with exit DNA extending from it. It then shows the high-resolution cryo-EM maps for RSC modules, and finally the structural model as a ribbon representation with subunits in different colours (colour code as in Fig. 1).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wagner, F.R., Dienemann, C., Wang, H. et al. Structure of SWI/SNF chromatin remodeller RSC bound to a nucleosome. Nature 579, 448–451 (2020). https://doi.org/10.1038/s41586-020-2088-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2088-0

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing