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.

Distinct roles of cohesin-SA1 and cohesin-SA2 in 3D chromosome organization

Abstract

Two variant cohesin complexes containing SMC1, SMC3, RAD21 and either SA1 (also known as STAG1) or SA2 (also known as STAG2) are present in all cell types. We report here their genomic distribution and specific contributions to genome organization in human cells. Although both variants are found at CCCTC-binding factor (CTCF) sites, a distinct population of the SA2-containing cohesin complexes (hereafter referred to as cohesin-SA2) localize to enhancers lacking CTCF, are linked to tissue-specific transcription and cannot be replaced by the SA1-containing cohesin complex (cohesin-SA1) when SA2 is absent, a condition that has been observed in several tumors. Downregulation of each of these variants has different consequences for gene expression and genome architecture. Our results suggest that cohesin-SA1 preferentially contributes to the stabilization of topologically associating domain boundaries together with CTCF, whereas cohesin-SA2 promotes cell-type-specific contacts between enhancers and promoters independently of CTCF. Loss of cohesin-SA2 rewires local chromatin contacts and alters gene expression. These findings provide insights into how cohesin mediates chromosome folding and establish a novel framework to address the consequences of mutations in cohesin genes in cancer.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: A large fraction of cohesin-SA2 localizes to enhancers independently of CTCF.
Fig. 2: Cohesin-SA2-only positions are enriched in cell-type-specific super-enhancers.
Fig. 3: SA2-specific changes in transcription are related to cell identity.
Fig. 4: Different behavior of cohesin in common and SA2-only positions.
Fig. 5: Cohesin-SA1 cannot occupy SA2-only sites.
Fig. 6: Distinct contribution of cohesin-SA1 and cohesin-SA2 to genome architecture.

References

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Zuin, J. et al. Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells. Proc. Natl. Acad. Sci. USA 111, 996–1001 (2014).

    Article  PubMed  CAS  Google Scholar 

  5. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Losada, A., Yokochi, T., Kobayashi, R. & Hirano, T. Identification and characterization of SA or Scc3p subunits in the Xenopus and human cohesin complexes. J. Cell Biol. 150, 405–416 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Canudas, S. & Smith, S. Differential regulation of telomere and centromere cohesion by the Scc3 homologs SA1 and SA2, respectively, in human cells. J. Cell Biol. 187, 165–173 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Remeseiro, S. et al. Cohesin-SA1 deficiency drives aneuploidy and tumorigenesis in mice due to impaired replication of telomeres. EMBO J. 31, 2076–2089 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. van der Lelij, P. et al. Synthetic lethality between the cohesin subunits STAG1 and STAG2 in diverse cancer contexts. eLife 6, e26980 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Cuadrado, A., Remeseiro, S., Gómez-López, G., Pisano, D. G. & Losada, A. The specific contributions of cohesin-SA1 to cohesion and gene expression: implications for cancer and development. Cell Cycle 11, 2233–2238 (2012).

    Article  PubMed  CAS  Google Scholar 

  11. De Koninck, M. & Losada, A. Cohesin mutations in cancer. Cold Spring Harb. Perspect. Med. 6, a026476 (2016).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  12. Balbás-Martínez, C. et al. Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy. Nat. Genet. 45, 1464–1469 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Solomon, D. A. et al. Frequent truncating mutations of STAG2 in bladder cancer. Nat. Genet. 45, 1428–1430 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Kon, A. et al. Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms. Nat. Genet. 45, 1232–1237 (2013).

    Article  PubMed  CAS  Google Scholar 

  15. Viny, A. D. et al. Dose-dependent role of the cohesin complex in normal and malignant hematopoiesis. J. Exp. Med. 212, 1819–1832 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Corces, M. R. & Corces, V. G. The three-dimensional cancer genome. Curr. Opin. Genet. Dev. 36, 1–7 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  18. Parelho, V. et al. Cohesins functionally associate with CTCF on mammalian chromosome arms. Cell 132, 422–433 (2008).

    Article  PubMed  CAS  Google Scholar 

  19. Rubio, E. D. et al. CTCF physically links cohesin to chromatin. Proc. Natl. Acad. Sci. USA 105, 8309–8314 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation center. Nature 485, 381–385 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Guo, Y. et al. CRISPR inversion of CTCF sites alters genome topology and enhancer–promoter function. Cell 162, 900–910 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Narendra, V. et al. CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation. Science 347, 1017–1021 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Haarhuis, J. H. I. et al. The cohesin release factor WAPL restricts chromatin loop extension. Cell 169, 693–707 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Sanborn, A. L. et al. Chromatin extrusion explains key features of loop and domain formation in wild-type and engineered genomes. Proc. Natl. Acad. Sci. USA 112, E6456–E6465 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Fudenberg, G. et al. Formation of chromosomal domains by loop extrusion. Cell Rep. 15, 2038–2049 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. de Wit, E. et al. CTCF binding polarity determines chromatin looping. Mol. Cell 60, 676–684 (2015).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Schmidt, D. et al. A CTCF-independent role for cohesin in tissue-specific transcription. Genome Res. 20, 578–588 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Faure, A. J. et al. Cohesin regulates tissue-specific expression by stabilizing highly occupied cis-regulatory modules. Genome Res. 22, 2163–2175 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Ernst, J. et al. Mapping and analysis of chromatin-state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Jang, W., Kim, T., Koo, J. S., Kim, S. K. & Lim, D. S. Mechanical-cue-induced YAP instructs SKP2-dependent cell cycle exit and oncogenic signaling. EMBO J. 36, 2510–2528 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  32. Whyte, W. A. et al. Master transcription factors and Mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Ballas, N., Grunseich, C., Lu, D. D., Speh, J. C. & Mandel, G. REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis. Cell 121, 645–657 (2005).

    Article  PubMed  CAS  Google Scholar 

  34. D’Alessio, A. C. et al. A systematic approach to identify candidate transcription factors that control cell identity. Stem Cell Rep. 5, 763–775 (2015).

    Article  CAS  Google Scholar 

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

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  36. Davidson, I. F. et al. Rapid movement and transcriptional relocalization of human cohesin on DNA. EMBO J. 35, 2671–2685 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  41. Wutz, G. et al. Topologically associating domains and chromatin loops depend on cohesin and are regulated by CTCF, WAPL and PDS5 proteins. EMBO J. 36, 3573–3599 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  42. Schwarzer, W. et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature 551, 51–56 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature 518, 331–336 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Flyamer, I. M. et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544, 110–114 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Stevens, T. J. et al. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature 544, 59–64 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Canudas, S. et al. Protein requirements for sister telomere association in human cells. EMBO J. 26, 4867–4878 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Murayama, Y., Samora, C. P., Kurokawa, Y., Iwasaki, H. & Uhlmann, F. Establishment of DNA–DNA interactions by the cohesin ring. Cell 172, 465–477 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Countryman, P. et al. Cohesin SA2 is a sequence-independent DNA-binding protein that recognizes DNA replication and repair intermediates. J. Biol. Chem. 293, 1054–1069 (2018).

    Article  PubMed  CAS  Google Scholar 

  49. Li, W. et al. Functional roles of enhancer RNAs for estrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Ing-Simmons, E. et al. Spatial enhancer clustering and regulation of enhancer-proximal genes by cohesin. Genome Res. 25, 504–513 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Mullenders, J. et al. Cohesin loss alters adult hematopoietic stem cell homeostasis, leading to myeloproliferative neoplasms. J. Exp. Med. 212, 1833–1850 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Carretero, M., Ruiz-Torres, M., Rodríguez-Corsino, M., Barthelemy, I. & Losada, A. PDS5B is required for cohesion establishment and aurora B accumulation at centromeres. EMBO J. 32, 2938–2949 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Gocke, C. B. & Yu, H. ZNF198 stabilizes the LSD1–CoREST–HDAC1 complex on chromatin through its MYM-type zinc fingers. PLoS One 3, e3255 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Méndez, J. & Stillman, B. Chromatin association of human origin recognition complex, CDC6 and minichromosome maintenance proteins during the cell cycle: assembly of pre-replication complexes in late mitosis. Mol. Cell. Biol. 20, 8602–8612 (2000).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Hu, B. et al. Biological chromodynamics: a general method for measuring protein occupancy across the genome by calibrating ChIP-seq. Nucleic Acids Res. 43, e132 (2015).

    PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Ramírez, F. et al. deepTools2: a next-generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44(W1), W160–W165 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Bailey, T. L. et al. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–W208 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Subramanian, A., Kuehn, H., Gould, J., Tamayo, P. & Mesirov, J. P. GSEA-P: a desktop application for gene set enrichment analysis. Bioinformatics 23, 3251–3253 (2007).

    Article  PubMed  CAS  Google Scholar 

  61. Serra, F. et al. Automatic analysis and 3D-modeling of Hi-C data using TADbit reveals structural features of the fly chromatin colors. PLoS Comput. Biol. 13, e1005665 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Wiśniewski, J. R., Zougman, A., Nagaraj, N. & Mann, M. Universal sample preparation method for proteome analysis. Nat. Methods 6, 359–362 (2009).

    Article  PubMed  CAS  Google Scholar 

  63. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  PubMed  CAS  Google Scholar 

  64. Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).

    Article  PubMed  CAS  Google Scholar 

  65. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Y. Cuartero and J. Quilez (4D Genome–CRG) for technical help with the Hi-C experiments, D. Rico (Newcastle University), F. X. Real (CNIO) and M. Manzanares (CNIC) for comments on the manuscript, T. Hirano (RIKEN) and H. Yu (UT Southwestern) for reagents, and M. Quintela (CNIO) for MCF10A cells. This work has been supported by the Spanish Ministry of Economy and Competitiveness and FEDER funds (grant no. BFU2013-48481-R (A.L.), BFU2016-79841-R (A.L.) and BFU2013-47736-P (M.A.M.-R.), fellowship no. BES-2014-069166 (M.D.K.), and Centro de Excelencia Severo Ochoa grant no. SEV-2015-0510 (to CNIO) and SEV-2012-0208 (to CRG), the European Research Council (FP7/2010-2015, ERC grant agreement 609989; M.A.M.-R.), the EU Horizon 2020 Research and Innovation Program (agreement 676556; M.A.M.-R.), the CERCA Programme–Generalitat de Catalunya (M.A.M.-R.) and the La Caixa Foundation (PhD fellowship to A.K.).

Author information

Authors and Affiliations

Authors

Contributions

A.C. and A.K. performed most of the experiments with technical help from M.R.-C.; M.D.K. performed the immunoprecipitation and salt-extraction experiments; F.L.D. and A.K. performed the Hi-C experiments; G.G.-L. analyzed the RNA-seq data; A.K. and D.G.-L. analyzed the ChIP–seq data; M.A.M.-R. analyzed the Hi-C data; A.C. and A.L. planned the project and wrote the manuscript with contributions from all of the authors.

Corresponding authors

Correspondence to Ana Cuadrado, Marc A. Marti-Renom or Ana Losada.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Cohesin variant abundance in different cell lines.

a, Immunoblots of total extracts from human primary cells (NHEK, normal human epithelial keratinocytes; PrEC, prostate epithelial cells; NHBE, normal human bronchial epithelial cells; NHA, normal human astrocytes; HCAEC, human coronary artery endothelial cells; SKMC, skeletal muscle cells; HMEC, human mammary epithelial cells; HUVEC, human umbilical vein endothelial cells; NHOst, normal human osteoblasts) and MCF10A cells. b, Known amounts of C-terminal fragments of SA1 and SA2 (around 50 kDa each) were run alongside the indicated fractions from MCF10A cells to assess relative abundance of cohesin-SA1 and cohesin-SA2 on chromatin (T, total cell extract; Cyt, cytosolic fraction; N, soluble nuclear fraction; Chr, chromatin-bound protein). The dotted line indicates that two parts of the film corresponding to the same exposure but different sizes have been pasted together. This was repeated with all cell lines used in a (data not shown). c, Quantification of the signals in the blots above was used to generate the histograms for total cohesin levels and the SA2:SA1 ratio in the same cell lines.

Supplementary Figure 2 Cohesin-SA2-only distribution around transcriptional regulators.

a, Motif-based analysis of the indicated positions in HMECs. E-values of the significantly enriched motifs and percentage of regions containing each motif for a given condition are indicated. The “TF” column contains the transcription factors with statistically significant binding (blue scale; right) to the identified motifs. b, Distribution of SMC1, SA1, SA2 and CTCF around Zmym2 and activated YAP (YAP-5SA) positions (±2.5 kb) defined by ChIP–seq in MCF10A cells.

Supplementary Figure 3 Cohesin-SA2 contributes to maintain cell identity.

Enrichment plots for genes within KEGG pathways specific for the hematopoietic system (“Hematopoietic cell lineage”) and the nervous system (“Neuroactive Ligand-Receptor interaction”) after downregulation of SA1, SA2 or CTCF. Both these pathways appear significantly upregulated in the siSA2 condition (NES > 0, in red; FWER q < 0.01). In the other two conditions, the gene expression changes are either not significant (FWER q value in gray) or in the opposite direction (NES < 0, in blue). The y axis shows the enrichment score (ES).

Supplementary Figure 4 Preferential accumulation of cohesin in SA2-only sites in Wapl-knockout cells.

Read density plots showing cohesin SMC1 distribution in wild-type (wt) and Wapl-knockout (KO) human HAP1 cells from ref. 23. Cohesin positions were classified into common and SA2-only based on their co-occurrence with CTCF in Wapl-knockout cells.

Supplementary Figure 5 Hi-C analyses in MCF10A cells after depletion of SA1 or SA2.

a, MCF10A cells depleted for SA1 or SA2 by RNAi were arrested in G1 by contact inhibition. Left, immunoblot analysis of the remaining levels of cohesin 72 h after transfection. Right, cell cycle profiles of MCF10A proliferating cells and of each of the samples used for Hi-C analysis (two replicas, r1 and r2, per condition). b, Hi-C normalized interaction maps for chromosome 15 at 100-kb resolution. Left, contact maps for r1 and r2 for each condition (siControl, siSA1 and siSA2). Middle, correlation between r1 and r2 of each condition of all data in the normalized interaction map for chromosome 15. Right, correlation between the Eigen values. c, Scatterplot of eigenvectors of the intrachromosomal interaction matrices indicated. Numbers within the plot show the percentage of bins that change compartment between replicates for each condition. d, TAD number, TAD border strength and TAD border conservation analysis as shown in Fig. 6c–e, including replicas. e, Hi-C interactions as a function of genomic distance averaged across the genome for the two replicas in the siSA1 and siSA2 conditions.

Supplementary Figure 6 Chromosomal interactions are differently affected by depletion of SA1 or SA2.

Chromosome 15 differential interaction map at 100-kb resolution of siSA1 (top) or siSA2 (down) versus siControl for the merged dataset. The panel is complemented by tracks of A/B compartment assignment based on PC1 Eigen values as well as gene density across the genome (middle of the interaction matrices). Replicate experiments are also shown on both sides.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6

Reporting Summary

Supplementary Table 1

Primer sequences

Supplementary Dataset 1

Uncropped blot images

Supplementary Dataset 2

ChIP–seq and RNA-seq datasets

Supplementary Dataset 3

Proteomic analyses of SA1 and SA2 immunoprecipitates

Supplementary Dataset 4

DEGs in siSA1 versus siControl

Supplementary Dataset 5

DEGs in siSA2 versus siControl

Supplementary Dataset 6

DEGs in siCTCF versus siControl

Supplementary Dataset 7

Hi-C data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kojic, A., Cuadrado, A., De Koninck, M. et al. Distinct roles of cohesin-SA1 and cohesin-SA2 in 3D chromosome organization. Nat Struct Mol Biol 25, 496–504 (2018). https://doi.org/10.1038/s41594-018-0070-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0070-4

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