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 cohesin subcomplex pinpoints direct shugoshin-Wapl antagonism in centromeric cohesion

Abstract

Orderly termination of sister-chromatid cohesion during mitosis is critical for accurate chromosome segregation. During prophase, mitotic kinases phosphorylate cohesin and its protector sororin, triggering Wapl-dependent cohesin release from chromosome arms. The shugoshin (Sgo1)–PP2A complex protects centromeric cohesin until its cleavage by separase at anaphase onset. Here, we report the crystal structure of a human cohesin subcomplex comprising SA2 and Scc1. Multiple HEAT repeats of SA2 form a dragon-shaped structure. Scc1 makes extensive contacts with SA2, with one binding hotspot. Sgo1 and Wapl compete for binding to a conserved site on SA2–Scc1. At this site, mutations of SA2 residues that disrupt Wapl binding bypass the Sgo1 requirement in cohesion protection. Thus, in addition to recruiting PP2A to dephosphorylate cohesin and sororin, Sgo1 physically shields cohesin from Wapl. This unexpected, direct antagonism between Sgo1 and Wapl augments centromeric cohesion protection.

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: Structure and binding interface of human SA2–Scc1.
Figure 2: Identification of a binding hotspot between SA2 and Scc1.
Figure 3: A conserved, functional Sgo1-binding site of SA2–Scc1.
Figure 4: Competition between Wapl and Sgo1 for cohesin binding.
Figure 5: Sgo1 prevents Wapl from accessing a functional site on cohesin.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

References

  1. Nasmyth, K. & Haering, C.H. Cohesin: its roles and mechanisms. Annu. Rev. Genet. 43, 525–558 (2009).

    Article  CAS  Google Scholar 

  2. Peters, J.M., Tedeschi, A. & Schmitz, J. The cohesin complex and its roles in chromosome biology. Genes Dev. 22, 3089–3114 (2008).

    Article  CAS  Google Scholar 

  3. Onn, I., Heidinger-Pauli, J.M., Guacci, V., Unal, E. & Koshland, D.E. Sister chromatid cohesion: a simple concept with a complex reality. Annu. Rev. Cell Dev. Biol. 24, 105–129 (2008).

    Article  CAS  Google Scholar 

  4. Wu, N. & Yu, H. The Smc complexes in DNA damage response. Cell Biosci. 2, 5 (2012).

    Article  Google Scholar 

  5. Wu, N. et al. Scc1 sumoylation by Mms21 promotes sister chromatid recombination through counteracting Wapl. Genes Dev. 26, 1473–1485 (2012).

    Article  CAS  Google Scholar 

  6. Merkenschlager, M. & Odom, D.T. CTCF and cohesin: linking gene regulatory elements with their targets. Cell 152, 1285–1297 (2013).

    Article  CAS  Google Scholar 

  7. Solomon, D.A. et al. Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science 333, 1039–1043 (2011).

    Article  CAS  Google Scholar 

  8. Remeseiro, S., Cuadrado, A. & Losada, A. Cohesin in development and disease. Development 140, 3715–3718 (2013).

    Article  CAS  Google Scholar 

  9. Nasmyth, K. Cohesin: a catenase with separate entry and exit gates? Nat. Cell Biol. 13, 1170–1177 (2011).

    Article  CAS  Google Scholar 

  10. Sherwood, R., Takahashi, T.S. & Jallepalli, P.V. Sister acts: coordinating DNA replication and cohesion establishment. Genes Dev. 24, 2723–2731 (2010).

    Article  CAS  Google Scholar 

  11. Shintomi, K. & Hirano, T. Sister chromatid resolution: a cohesin releasing network and beyond. Chromosoma 119, 459–467 (2010).

    Article  Google Scholar 

  12. Ciosk, R. et al. Cohesin's binding to chromosomes depends on a separate complex consisting of Scc2 and Scc4 proteins. Mol. Cell 5, 243–254 (2000).

    Article  CAS  Google Scholar 

  13. Gillespie, P.J. & Hirano, T. Scc2 couples replication licensing to sister chromatid cohesion in Xenopus egg extracts. Curr. Biol. 14, 1598–1603 (2004).

    Article  CAS  Google Scholar 

  14. Watrin, E. et al. Human Scc4 is required for cohesin binding to chromatin, sister-chromatid cohesion, and mitotic progression. Curr. Biol. 16, 863–874 (2006).

    Article  CAS  Google Scholar 

  15. Tonkin, E.T., Wang, T.J., Lisgo, S., Bamshad, M.J. & Strachan, T. NIPBL, encoding a homolog of fungal Scc2-type sister chromatid cohesion proteins and fly Nipped-B, is mutated in Cornelia de Lange syndrome. Nat. Genet. 36, 636–641 (2004).

    Article  CAS  Google Scholar 

  16. Losada, A., Yokochi, T. & Hirano, T. Functional contribution of Pds5 to cohesin-mediated cohesion in human cells and Xenopus egg extracts. J. Cell Sci. 118, 2133–2141 (2005).

    Article  CAS  Google Scholar 

  17. Gandhi, R., Gillespie, P.J. & Hirano, T. Human Wapl is a cohesin-binding protein that promotes sister-chromatid resolution in mitotic prophase. Curr. Biol. 16, 2406–2417 (2006).

    Article  CAS  Google Scholar 

  18. Kueng, S. et al. Wapl controls the dynamic association of cohesin with chromatin. Cell 127, 955–967 (2006).

    Article  CAS  Google Scholar 

  19. Rankin, S., Ayad, N.G. & Kirschner, M.W. Sororin, a substrate of the anaphase-promoting complex, is required for sister chromatid cohesion in vertebrates. Mol. Cell 18, 185–200 (2005).

    Article  CAS  Google Scholar 

  20. Lafont, A.L., Song, J. & Rankin, S. Sororin cooperates with the acetyltransferase Eco2 to ensure DNA replication-dependent sister chromatid cohesion. Proc. Natl. Acad. Sci. USA 107, 20364–20369 (2010).

    Article  CAS  Google Scholar 

  21. Nishiyama, T. et al. Sororin mediates sister chromatid cohesion by antagonizing Wapl. Cell 143, 737–749 (2010).

    Article  CAS  Google Scholar 

  22. Hauf, S. et al. Dissociation of cohesin from chromosome arms and loss of arm cohesion during early mitosis depends on phosphorylation of SA2. PLoS Biol. 3, e69 (2005).

    Article  Google Scholar 

  23. Dreier, M.R., Bekier, M.E. II & Taylor, W.R. Regulation of sororin by Cdk1-mediated phosphorylation. J. Cell Sci. 124, 2976–2987 (2011).

    Article  CAS  Google Scholar 

  24. Nishiyama, T., Sykora, M.M., Huis In 't Veld, P.J., Mechtler, K. & Peters, J.M. Aurora B and Cdk1 mediate Wapl activation and release of acetylated cohesin from chromosomes by phosphorylating Sororin. Proc. Natl. Acad. Sci. USA 110, 13404–13409 (2013).

    Article  CAS  Google Scholar 

  25. Kitajima, T.S. et al. Shugoshin collaborates with protein phosphatase 2A to protect cohesin. Nature 441, 46–52 (2006).

    Article  CAS  Google Scholar 

  26. Riedel, C.G. et al. Protein phosphatase 2A protects centromeric sister chromatid cohesion during meiosis I. Nature 441, 53–61 (2006).

    Article  CAS  Google Scholar 

  27. Tang, Z. et al. PP2A is required for centromeric localization of Sgo1 and proper chromosome segregation. Dev. Cell 10, 575–585 (2006).

    Article  CAS  Google Scholar 

  28. Liu, H., Rankin, S. & Yu, H. Phosphorylation-enabled binding of SGO1–PP2A to cohesin protects sororin and centromeric cohesion during mitosis. Nat. Cell Biol. 15, 40–49 (2013).

    Article  CAS  Google Scholar 

  29. Foley, E.A. & Kapoor, T.M. Microtubule attachment and spindle assembly checkpoint signalling at the kinetochore. Nat. Rev. Mol. Cell Biol. 14, 25–37 (2013).

    Article  CAS  Google Scholar 

  30. Jia, L., Kim, S. & Yu, H. Tracking spindle checkpoint signals from kinetochores to APC/C. Trends Biochem. Sci. 38, 302–311 (2013).

    Article  CAS  Google Scholar 

  31. Lee, J. et al. Unified mode of centromeric protection by shugoshin in mammalian oocytes and somatic cells. Nat. Cell Biol. 10, 42–52 (2008).

    Article  CAS  Google Scholar 

  32. Liu, H., Jia, L. & Yu, H. Phospho-H2A and cohesin specify distinct tension-regulated Sgo1 pools at kinetochores and inner centromeres. Curr. Biol. 23, 1927–1933 (2013).

    Article  CAS  Google Scholar 

  33. Tang, Z., Shu, H., Oncel, D., Chen, S. & Yu, H. Phosphorylation of Cdc20 by Bub1 provides a catalytic mechanism for APC/C inhibition by the spindle checkpoint. Mol. Cell 16, 387–397 (2004).

    Article  CAS  Google Scholar 

  34. McGuinness, B.E., Hirota, T., Kudo, N.R., Peters, J.M. & Nasmyth, K. Shugoshin prevents dissociation of cohesin from centromeres during mitosis in vertebrate cells. PLoS Biol. 3, e86 (2005).

    Article  Google Scholar 

  35. Murayama, Y. & Uhlmann, F. Biochemical reconstitution of topological DNA binding by the cohesin ring. Nature 505, 367–371 (2014).

    Article  CAS  Google Scholar 

  36. Shintomi, K. & Hirano, T. Releasing cohesin from chromosome arms in early mitosis: opposing actions of Wapl-Pds5 and Sgo1. Genes Dev. 23, 2224–2236 (2009).

    Article  CAS  Google Scholar 

  37. Ivanovic, T., Choi, J.L., Whelan, S.P., van Oijen, A.M. & Harrison, S.C. Influenza-virus membrane fusion by cooperative fold-back of stochastically induced hemagglutinin intermediates. Elife 2, e00333 (2013).

    Article  Google Scholar 

  38. Ouyang, Z. et al. Structure of the human cohesin inhibitor Wapl. Proc. Natl. Acad. Sci. USA 110, 11355–11360 (2013).

    Article  CAS  Google Scholar 

  39. Tedeschi, A. et al. Wapl is an essential regulator of chromatin structure and chromosome segregation. Nature 501, 564–568 (2013).

    Article  CAS  Google Scholar 

  40. Haarhuis, J.H. et al. WAPL-mediated removal of cohesin protects against segregation errors and aneuploidy. Curr. Biol. 23, 2071–2077 (2013).

    Article  CAS  Google Scholar 

  41. Yu, H. Chromosome biology: Wapl spreads its wings. Curr. Biol. 23, R923–R925 (2013).

    Article  CAS  Google Scholar 

  42. Daum, J.R. et al. Cohesion fatigue induces chromatid separation in cells delayed at metaphase. Curr. Biol. 21, 1018–1024 (2011).

    Article  CAS  Google Scholar 

  43. Xu, Z. et al. Structure and function of the PP2A-shugoshin interaction. Mol. Cell 35, 426–441 (2009).

    Article  CAS  Google Scholar 

  44. Buheitel, J. & Stemmann, O. Prophase pathway-dependent removal of cohesin from human chromosomes requires opening of the Smc3-Scc1 gate. EMBO J. 32, 666–676 (2013).

    Article  CAS  Google Scholar 

  45. Eichinger, C.S., Kurze, A., Oliveira, R.A. & Nasmyth, K. Disengaging the Smc3/kleisin interface releases cohesin from Drosophila chromosomes during interphase and mitosis. EMBO J. 32, 656–665 (2013).

    Article  CAS  Google Scholar 

  46. Chatterjee, A., Zakian, S., Hu, X.W. & Singleton, M.R. Structural insights into the regulation of cohesion establishment by Wpl1. EMBO J. 32, 677–687 (2013).

    Article  CAS  Google Scholar 

  47. Kitajima, T.S., Kawashima, S.A. & Watanabe, Y. The conserved kinetochore protein shugoshin protects centromeric cohesion during meiosis. Nature 427, 510–517 (2004).

    Article  CAS  Google Scholar 

  48. Katis, V.L., Galova, M., Rabitsch, K.P., Gregan, J. & Nasmyth, K. Maintenance of cohesin at centromeres after meiosis I in budding yeast requires a kinetochore-associated protein related to MEI-S332. Curr. Biol. 14, 560–572 (2004).

    Article  CAS  Google Scholar 

  49. Hauf, S., Waizenegger, I.C. & Peters, J.M. Cohesin cleavage by separase required for anaphase and cytokinesis in human cells. Science 293, 1320–1323 (2001).

    Article  CAS  Google Scholar 

  50. Otwinowski, Z. & Minor, W. Processing X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  52. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  Google Scholar 

Download references

Acknowledgements

We thank C. Brautigam for assistance with ITC. Use of Argonne National Laboratory Structural Biology Center beamlines at the Advanced Photon Source was supported by the US Department of Energy under contract DE-AC02-06CH11357. H.Y. is supported as an investigator of the Howard Hughes Medical Institute. This work is supported by grants from the Cancer Prevention and Research Institute of Texas (RP110465-P3 to H.Y.) and the Welch Foundation (I-1441 to H.Y.).

Author information

Authors and Affiliations

Authors

Contributions

K.H. crystallized SA2–Scc1 and determined its structure with the help of Z.C. and D.R.T. and performed the in vitro binding assays with SA2 and Scc1 mutants. G.Z. performed the functional cellular assays. Q.Q. performed the cohesin binding assay that showed competition between Sgo1 and Wapl and the in vitro binding assay between SA2–Scc1 and Wapl. Z.O. first observed competition between Sgo1 and Wapl in cohesin binding. H.L. performed the metaphase spread assay of sororin 9A–expressing cells depleted of Sgo1. H.Y. supervised the project and wrote the paper with input from all authors.

Corresponding author

Correspondence to Hongtao Yu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Mutational analysis of the SA2-Scc1 interaction in vitro and in human cells.

(a) Autoradiograph (top) and Coomassie stained gel (bottom) of 35S-labeled Myc-SA2 proteins (input) and the same proteins bound to GST or GST-Scc1 beads. WT, wild type. (b) Quantification of the in vitro binding assays in a. The binding intensities were normalized to the amount of input for each SA2 protein. Error bars, s.d. (n = 3, independent experiments). The Y331A, Y479A, K1009E L1010A mutants were tested twice. Only the means were shown for these samples. (c) Autoradiograph (top) and Coomassie stained gel (bottom) of 35S-labeled Scc1-Myc proteins (input) and the same proteins bound to GST or GST-SA2 beads. WT, wild type. (d) Quantification of the in vitro binding assays in c. The binding intensities were normalized to the amount of input for each Scc1 protein. Error bars, s.d. (n = 3, independent experiments). (e) Anti-Myc or anti-GFP immunoblots of lysates and anti-Myc IP of HeLa cells co-transfected with plasmids encoding GFP-SA2 and the indicated Myc-Scc1 proteins. WT, wild type.

Supplementary Figure 2 Identification of a Sgo1-binding site on SA2–Scc1.

(a,b) Quantification of normalized intensities of the indicated 35S-labeled SA2–Scc1 proteins bound to beads coupled to phospho-T346 Sgo1 peptides. WT, wild type. Error bars, s.d. (n = 3, independent experiments). (c) Anti-Myc, anti-GFP, and anti-Sgo1 blots of lgG and anti-GFP IP from GFP-Sgo1-expressing HeLa cells co-transfected with Scc1-Myc and the indicated Myc-SA2 plasmids. WT, wild type. (d) Anti-Myc, anti-SA2, and anti-tubulin blots of lysates of HeLa cells transfected with the indicated siRNAs and plasmids. WT, wild type. (e) Quantification of mitotic indices (defined as MPM2-positive, 4N cells) of cells in d. Error bars, s.d. (n = 7, independent experiments).

Supplementary Figure 3 The conserved FVHRYRD motif of SA2 is not required for cohesin loading in human cells.

(a) Cartoon diagram of the structure of human SA2–Scc1, with SA2 colored blue and Scc1 colored pink. The conserved SA2–Scc1 regions implicated in binding the cohesin loader Scc2–Scc4 are colored yellow. The MES molecule bound near the SA2–Scc1 interface is shown in sticks. The chemical structure of MES is shown in the lower right corner. (b) A zoomed-in view of the MES-binding site, with the kicked OMIT map of electron density around MES shown at a contour level of 3σ. (c) DAPI (blue in merge), anti-GFP (red in merge), and anti-tubulin (green in merge) staining of telophase HeLa cells transfected with the indicated GFP-SA2 plasmids and siRNAs. WT, wild type. Scale bar, 10 μm. (d) Quantification of the anti-GFP staining intensities of cells in c. Each dot in the graph represents a single cell (Mock, n = 12; Vector, n = 22; WT, n = 48; Y297A, n = 18; R298E, n = 46; D793K, n = 40). The horizontal bars indicate the means.

Supplementary Figure 4 Identification of a Wapl-binding site on SA2–Scc1.

(a) Anti-Wapl and anti-tubulin blots of lysates of HeLa cells transfected with the indicated siRNAs with or without increasing amounts of the indicated GFP-Wapl plasmids. WT, wild type. The positions of the endogenous and GFP-Wapl are labeled. (b) Quantification of mitotic indices (defined as MPM2-positive, 4N cells) of cells in a. Error bars, range (n = 2, independent experiments). (c) Autoradiograph (top) and Coomassie stained gel (bottom) of 35S-labeled Myc-SA2–Scc1 proteins (input) and the same proteins bound to beads containing GST or increasing amounts of GST-Wapl-M. WT, wild type. (d) Quantification of the in vitro binding assays in c. The binding intensities were normalized to the amount of each input. Error bars, range (n = 2, independent experiments). The K290E and D326K mutants were only tested once.

Supplementary Figure 5 Expression of Wapl binding–deficient SA2 mutants bypasses Sgo1 requirement in cohesion protection and rescues cohesion fatigue.

(a) Anti-SA2, anti-Myc, and anti-tubulin blots of lysates of HeLa cells transfected with the indicated siRNA and plasmids. WT, wild type. (b) Four major types (I-IV) of metaphase spreads of cells in a, stained with DAPI (blue) and the kinetochore marker CREST (red). Selected sister chromatids were magnified and shown in insets. Scale bar, 5 μm. (c) Quantification of the percentage of cells in a with type III and IV chromosome morphologies as in b. Error bars, range (n = 2, independent experiments). The K330E mutant was tested only once. (d) Quantification of the percentages of mitotic HeLa cells (transfected with the indicated Myc-SA2 plasmids, arrested in nocodazole, and released into medium containing MG132 for 2 hrs) that had types III/IV chromosome morphology. WT, wild type. Error bars, s.d. for mock, WT, D326K (n = 3, independent experiments); range for K330E and Y331A (n = 2, independent experiments).

Supplementary Figure 6 Uncropped original images of gels, autoradiographs, and blots presented in the main figures of this study.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 1254 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hara, K., Zheng, G., Qu, Q. et al. Structure of cohesin subcomplex pinpoints direct shugoshin-Wapl antagonism in centromeric cohesion. Nat Struct Mol Biol 21, 864–870 (2014). https://doi.org/10.1038/nsmb.2880

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2880

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