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.

  • Letter
  • Published:

Wapl is an essential regulator of chromatin structure and chromosome segregation

Abstract

Mammalian genomes contain several billion base pairs of DNA that are packaged in chromatin fibres. At selected gene loci, cohesin complexes have been proposed to arrange these fibres into higher-order structures1,2,3,4,5,6,7, but how important this function is for determining overall chromosome architecture and how the process is regulated are not well understood. Using conditional mutagenesis in the mouse, here we show that depletion of the cohesin-associated protein Wapl8,9 stably locks cohesin on DNA, leads to clustering of cohesin in axial structures, and causes chromatin condensation in interphase chromosomes. These findings reveal that the stability of cohesin–DNA interactions is an important determinant of chromatin structure, and indicate that cohesin has an architectural role in interphase chromosome territories. Furthermore, we show that regulation of cohesin–DNA interactions by Wapl is important for embryonic development, expression of genes such as c-myc (also known as Myc), and cell cycle progression. In mitosis, Wapl-mediated release of cohesin from DNA is essential for proper chromosome segregation and protects cohesin from cleavage by the protease separase, thus enabling mitotic exit in the presence of functional cohesin complexes.

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: Wapl depletion reveals arrangement of cohesin in axial chromosomal domains.
Figure 2: Wapl controls chromatin structure by regulating cohesin–DNA interactions.
Figure 3: Wapl is essential for cell cycle progression and proper chromosome segregation.
Figure 4: The prophase pathway of cohesin dissociation protects cohesin from cleavage by separase.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

Data deposits

ChiP-seq and microarray data have been deposited in the GEO database (http://www.ncbi.nlm.nih.gov/geo) under accession code GSE41603.

References

  1. Hadjur, S. et al. Cohesins form chromosomal cis-interactions at the developmentally regulated IFNG locus. Nature 460, 410–413 (2009)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  2. Mishiro, T. et al. Architectural roles of multiple chromatin insulators at the human apolipoprotein gene cluster. EMBO J. 28, 1234–1245 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nativio, R. et al. Cohesin is required for higher-order chromatin conformation at the imprinted IGF2–H19 locus. PLoS Genet. 5, e1000739 (2009)

    Article  PubMed  PubMed Central  Google Scholar 

  4. Kagey, M. H. et al. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467, 430–435 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  5. Seitan, V. C. et al. A role for cohesin in T-cell-receptor rearrangement and thymocyte differentiation. Nature 476, 467–471 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  6. DeMare, L. E. et al. The genomic landscape of cohesin-associated chromatin interactions. Genome Res. http://dx.doi.org/10.1101/gr.156570.113 (23 May 2013)

  7. Phillips-Cremins, J. E. et al. Architectural protein subclasses shape 3D organization of genomes during lineage commitment. Cell 153, 1281–1295 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Haering, C. H., Farcas, A. M., Arumugam, P., Metson, J. & Nasmyth, K. The cohesin ring concatenates sister DNA molecules. Nature 454, 297–301 (2008)

    Article  CAS  ADS  PubMed  Google Scholar 

  12. Chan, K. L. et al. Cohesin’s DNA exit gate is distinct from its entrance gate and is regulated by acetylation. Cell 150, 961–974 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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  PubMed  PubMed Central  Google Scholar 

  14. Uhlmann, F., Lottspeich, F. & Nasmyth, K. Sister-chromatid separation at anaphase onset is promoted by cleavage of the cohesin subunit Scc1. Nature 400, 37–42 (1999)

    Article  CAS  ADS  PubMed  Google Scholar 

  15. Waizenegger, I. C., Hauf, S., Meinke, A. & Peters, J. M. Two distinct pathways remove mammalian cohesin from chromosome arms in prophase and from centromeres in anaphase. Cell 103, 399–410 (2000)

    Article  CAS  PubMed  Google Scholar 

  16. 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  ADS  PubMed  Google Scholar 

  17. Kumada, K. et al. The selective continued linkage of centromeres from mitosis to interphase in the absence of mammalian separase. J. Cell Biol. 172, 835–846 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wirth, K. G. et al. Separase: a universal trigger for sister chromatid disjunction but not chromosome cycle progression. J. Cell Biol. 172, 847–860 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Seitan, V. C. & Merkenschlager, M. Cohesin and chromatin organisation. Curr. Opin. Genet. Dev. 22, 93–100 (2012)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  21. Schmitz, J., Watrin, E., Lenart, P., Mechtler, K. & Peters, J. M. Sororin is required for stable binding of cohesin to chromatin and for sister chromatid cohesion in interphase. Curr. Biol. 17, 630–636 (2007)

    Article  CAS  PubMed  Google Scholar 

  22. Wang, L. H., Mayer, B., Stemmann, O. & Nigg, E. A. Centromere DNA decatenation depends on cohesin removal and is required for mammalian cell division. J. Cell Sci. 123, 806–813 (2010)

    Article  CAS  PubMed  Google Scholar 

  23. Farcas, A. M., Uluocak, P., Helmhart, W. & Nasmyth, K. Cohesin’s concatenation of sister DNAs maintains their intertwining. Mol. Cell 44, 97–107 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chan, K. L. & Hickson, I. D. New insights into the formation and resolution of ultra-fine anaphase bridges. Semin. Cell Dev. Biol. 22, 906–912 (2011)

    Article  CAS  PubMed  Google Scholar 

  25. Sun, Y. et al. Separase is recruited to mitotic chromosomes to dissolve sister chromatid cohesion in a DNA-dependent manner. Cell 137, 123–132 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Vernì, F., Gandhi, R., Goldberg, M. L. & Gatti, M. Genetic and molecular analysis of wings apart-like (wapl), a gene controlling heterochromatin organization in Drosophila melanogaster. Genetics 154, 1693–1710 (2000)

    PubMed  PubMed Central  Google Scholar 

  27. Guacci, V. & Koshland, D. Cohesin-independent segregation of sister chromatids in budding yeast. Mol. Biol. Cell 23, 729–739 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lopez-Serra, L., Lengronne, A., Borges, V., Kelly, G. & Uhlmann, F. Budding yeast Wapl controls sister chromatid cohesion maintenance and chromosome condensation. Curr. Biol. 23, 64–69 (2013)

    Article  CAS  PubMed  Google Scholar 

  29. Pezzi, N. et al. STAG3, a novel gene encoding a protein involved in meiotic chromosome pairing and location of STAG3-related genes flanking the Williams– Beuren syndrome deletion. FASEB J. 14, 581–592 (2000)

    Article  CAS  PubMed  Google Scholar 

  30. Ono, T. et al. Differential contributions of condensin I and condensin II to mitotic chromosome architecture in vertebrate cells. Cell 115, 109–121 (2003)

    Article  CAS  PubMed  Google Scholar 

  31. Rodríguez, C. I. et al. High-efficiency deleter mice show that FLPe is an alternative to Cre-loxP. Nature Genet. 25, 139–140 (2000)

    PubMed  Google Scholar 

  32. Tallquist, M. D. & Soriano, P. Epiblast-restricted Cre expression in MORE mice: a tool to distinguish embryonic vs. extra-embryonic gene function. Genesis 26, 113–115 (2000)

    Article  CAS  PubMed  Google Scholar 

  33. Seibler, J. et al. Rapid generation of inducible mouse mutants. Nucleic Acids Res. 31, e12 (2003)

    Article  PubMed  PubMed Central  Google Scholar 

  34. Hadjantonakis, A. K. & Papaioannou, V. E. Dynamic in vivo imaging and cell tracking using a histone fluorescent protein fusion in mice. BMC Biotechnol. 4, 33 (2004)

    Article  PubMed  PubMed Central  Google Scholar 

  35. Poser, I. et al. BAC TransgeneOmics: a high-throughput method for exploration of protein function in mammals. Nature Methods 5, 409–415 (2008)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  36. Hauf, S. et al. The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore-microtubule attachment and in maintaining the spindle assembly checkpoint. J. Cell Biol. 161, 281–294 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  38. Schermelleh, L., Solovei, I., Zink, D. & Cremer, T. Two-color fluorescence labeling of early and mid-to-late replicating chromatin in living cells. Chromosome Res. 9, 77–80 (2001)

    Article  CAS  PubMed  Google Scholar 

  39. Zink, D. et al. Structure and dynamics of human interphase chromosome territories in vivo. Hum. Genet. 102, 241–251 (1998)

    Article  CAS  PubMed  Google Scholar 

  40. Stedman, W. et al. Cohesins localize with CTCF at the KSHV latency control region and at cellular c-myc and H19/Igf2 insulators. EMBO J. 27, 654–666 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wendt, K. S. et al. Cohesin mediates transcriptional insulation by CCCTC-binding factor. Nature 451, 796–801 (2008)

    Article  CAS  ADS  PubMed  Google Scholar 

  42. Bugler, B., Schmitt, E., Aressy, B. & Ducommun, B. Unscheduled expression of CDC25B in S-phase leads to replicative stress and DNA damage. Mol. Cancer 9, 29 (2010)

    Article  PubMed  PubMed Central  Google Scholar 

  43. Cerqueira, A. et al. Overall Cdk activity modulates the DNA damage response in mammalian cells. J. Cell Biol. 187, 773–780 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gerlich, D., Koch, B., Dupeux, F., Peters, J. M. & Ellenberg, J. Live-cell imaging reveals a stable cohesin–chromatin interaction after but not before DNA replication. Curr. Biol. 16, 1571–1578 (2006)

    Article  CAS  PubMed  Google Scholar 

  45. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nature Methods 9, 671–675 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We dedicate this paper to the memory of B. Peters, who performed the first experiments on Wapl in our laboratory. We are grateful to K. Aumayr, O. F.-Capetillo, T. Hoffmann, M. E. Idarraga-Amado, S. Kueng, T. Kulcsar, M. Leeb, P. Pasierbek, D. Santamaría, G. Schmauss, A. Souabni, H. Tkadletz, K. Wendt and members of the Peters laboratory for discussions and assistance, J. Hutchins for suggesting the term vermicelli, K. Nasmyth for the separase mouse model, and M. Barbacid, T. Cremer, T. Hirano, T. Jenuwein, M. Malumbres and J. Zuber for reagents. T.N. was supported by the European Molecular Biology Organization (EMBO) and the Japanese Society for the Promotion of Science (JSPS). S.H. was supported by funds from the Agence National de la Recherche (JCJC-SVSE2-2011, ChromaTranscript project) and the European Union (FP7-PEOPLE-2011-CIG, ChromaTranscript project). D.A.C. was supported by MFPL VIPS Program (BMWF and City of Vienna). A.V. acknowledges financial support by the Vienna Science and Technology Fund (WWTF) project VRG10-11, the Research Platform Quantum Phenomena and Nanoscale Biological Systems (QuNaBioS) and by Boehringer Ingelheim. Research in the laboratory of J.-M.P. is supported by Boehringer Ingelheim, the Austrian Science Fund (FWF special research program SFB F34 ‘Chromosome Dynamics’, and Wittgenstein award Z196-B20), the Austrian Research Promotion Agency (FFG, Laura Bassi Center for Optimized Structural Studies), the Vienna Science and Technology Fund (WWTF LS09-13), and the European Community’s Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 241548 (MitoSys).

Author information

Authors and Affiliations

Authors

Contributions

Experiments were designed and data interpreted by A.T., G.W., S.H., M.J., J.E. and J.-M.P. A.T. generated Waplfl embryonic stem cells, established and maintained the mouse colony required for this study, and carried out chromosome segregation, transcriptome and ChIP-seq studies. A.T. and G.W. analysed cohesin localization by IFM, and cleavage of Scc1 in anaphase MEFs. G.W. generated MEFs expressing Scc1–LAP and Scc1–9myc, and carried out cohesin IFM experiments in telophase and G1-phase MEFs. G.W. and V.B. performed chromatin fractionation experiments. G.W. and T.N. performed cleavage assays of Scc1 with Xenopus egg extracts. A.T., G.W. and W.T analysed cell cycle progression of MEFs. S.H. performed DNA granularity analysis and Cy3-dUTP labelling of DNA. M.J. carried out bioinformatic analyses. A.Wue. performed cohesin FRAP experiments. E.S. performed FISH experiments. A.Wut. supervised the work of A.T. with embryonic stem cells, and participated in designing the Wapl targeting strategy. I.F.D. performed replication assays with Xenopus egg extracts. D.A.C. performed live imaging of cohesin vermicelli under the guidance of A.V. A.T. and J.-M.P. wrote the manuscript.

Corresponding author

Correspondence to Jan-Michael Peters.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-18, Supplementary Methods, additional references and primer sequences for Supplementary figure 11. (PDF 2113 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tedeschi, A., Wutz, G., Huet, S. et al. Wapl is an essential regulator of chromatin structure and chromosome segregation. Nature 501, 564–568 (2013). https://doi.org/10.1038/nature12471

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12471

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