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:

Cohesin is positioned in mammalian genomes by transcription, CTCF and Wapl

Abstract

Mammalian genomes are spatially organized by CCCTC-binding factor (CTCF) and cohesin into chromatin loops1,2 and topologically associated domains3,4,5,6, which have important roles in gene regulation1,2,4,5,7 and recombination7,8,9. By binding to specific sequences10, CTCF defines contact points for cohesin-mediated long-range chromosomal cis-interactions1,2,4,5,6,7,11. Cohesin is also present at these sites12,13, but has been proposed to be loaded onto DNA elsewhere14,15 and to extrude chromatin loops until it encounters CTCF bound to DNA16,17,18,19. How cohesin is recruited to CTCF sites, according to this or other models, is unknown. Here we show that the distribution of cohesin in the mouse genome depends on transcription, CTCF and the cohesin release factor Wings apart-like (Wapl). In CTCF-depleted fibroblasts, cohesin cannot be properly recruited to CTCF sites but instead accumulates at transcription start sites of active genes, where the cohesin-loading complex is located14,15. In the absence of both CTCF and Wapl, cohesin accumulates in up to 70 kilobase-long regions at 3′-ends of active genes, in particular if these converge on each other. Changing gene expression modulates the position of these ‘cohesin islands’. These findings indicate that transcription can relocate mammalian cohesin over long distances on DNA, as previously reported for yeast cohesin20,21,22,23, that this translocation contributes to positioning cohesin at CTCF sites, and that active genes can be freed from cohesin either by transcription-mediated translocation or by Wapl-mediated release.

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

Figure 1: Cohesin distribution in WT, Ctcf, Smc3 and Wapl KO MEFs.
Figure 2: Cohesin redistribution to transcriptional start sites in Ctcf KO MEFs.
Figure 3: Cohesin accumulation at sites of convergent transcription in Ctcf Wapl DKO MEFs.
Figure 4: Transcription is required for the generation of cohesin islands.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

    Article  ADS  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  ADS  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  ADS  CAS  Google Scholar 

  5. Sofueva, S. et al. Cohesin-mediated interactions organize chromosomal domain architecture. EMBO J. 32, 3119–3129 (2013)

    Article  CAS  Google Scholar 

  6. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  8. Guo, C. et al. CTCF-binding elements mediate control of V(D)J recombination. Nature 477, 424–430 (2011)

    Article  ADS  CAS  Google Scholar 

  9. Medvedovic, J. et al. Flexible long-range loops in the VH gene region of the Igh locus facilitate the generation of a diverse antibody repertoire. Immunity 39, 229–244 (2013)

    Article  CAS  Google Scholar 

  10. Kim, T. H. et al. Analysis of the vertebrate insulator protein CTCF-binding sites in the human genome. Cell 128, 1231–1245 (2007)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  15. Zuin, J. et al. A cohesin-independent role for NIPBL at promoters provides insights in CdLS. PLoS Genet. 10, e1004153 (2014)

    Article  Google Scholar 

  16. Nasmyth, K. Disseminating the genome: joining, resolving, and separating sister chromatids during mitosis and meiosis. Annu. Rev. Genet. 35, 673–745 (2001)

    Article  CAS  Google Scholar 

  17. Nichols, M. H. & Corces, V. G. A CTCF code for 3D genome architecture. Cell 162, 703–705 (2015)

    Article  CAS  Google Scholar 

  18. 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  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. Glynn, E. F. et al. Genome-wide mapping of the cohesin complex in the yeast Saccharomyces cerevisiae. PLoS Biol. 2, E259 (2004)

    Article  Google Scholar 

  21. Lengronne, A. et al. Cohesin relocation from sites of chromosomal loading to places of convergent transcription. Nature 430, 573–578 (2004)

    Article  ADS  CAS  Google Scholar 

  22. 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 

  23. Gullerova, M. & Proudfoot, N. J. Cohesin complex promotes transcriptional termination between convergent genes in S. pombe. Cell 132, 983–995 (2008)

    Article  CAS  Google Scholar 

  24. Heath, H. et al. CTCF regulates cell cycle progression of αβ T cells in the thymus. EMBO J. 27, 2839–2850 (2008)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  26. Revilla-i-Domingo, R. et al. The B-cell identity factor Pax5 regulates distinct transcriptional programmes in early and late B lymphopoiesis. EMBO J. 31, 3130–3146 (2012)

    Article  CAS  Google Scholar 

  27. Schaaf, C. A. et al. Genome-wide control of RNA polymerase II activity by cohesin. PLoS Genet. 9, e1003382 (2013)

    Article  CAS  Google Scholar 

  28. Izumi, K. et al. Germline gain-of-function mutations in AFF4 cause a developmental syndrome functionally linking the super elongation complex and cohesin. Nature Genet. 47, 338–344 (2015)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Stigler, J., Çamdere, G. Ö., Koshland, D. E. & Greene, E. C. Single-molecule imaging reveals a collapsed conformational state for DNA-bound cohesin. Cell Reports 15, 988–998 (2016)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Uhlmann, F. & Nasmyth, K. Cohesion between sister chromatids must be established during DNA replication. Curr. Biol. 8, 1095–1101 (1998)

    Article  CAS  Google Scholar 

  33. Tachibana-Konwalski, K. et al. Rec8-containing cohesin maintains bivalents without turnover during the growing phase of mouse oocytes. Genes Dev. 24, 2505–2516 (2010)

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  Google Scholar 

  36. Sambrook, J & Russell, D. W. Molecular Cloning: A Laboratory Manual Vol. 3, A9.25–A9.34 (Cold Spring Harbor Laboratory Press, 2001)

    Google Scholar 

  37. Core, L. J., Waterfall, J. J. & Lis, J. T. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322, 1845–1848 (2008)

    Article  ADS  CAS  Google Scholar 

  38. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009)

    Article  Google Scholar 

  39. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009)

    Article  CAS  Google Scholar 

  40. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010)

    Article  CAS  Google Scholar 

  41. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010)

    Article  CAS  Google Scholar 

  42. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012)

    Article  CAS  Google Scholar 

  43. Benjamini, Y . & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995)

    MathSciNet  MATH  Google Scholar 

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

    Article  Google Scholar 

  45. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010)

    Article  CAS  Google Scholar 

  46. Aszódi, A. MULTOVL: fast multiple overlaps of genomic regions. Bioinformatics 28, 3318–3319 (2012)

    Article  Google Scholar 

  47. Shin, H., Liu, T., Manrai, A. K. & Liu, X. S. CEAS: cis-regulatory element annotation system. Bioinformatics 25, 2605–2606 (2009)

    Article  CAS  Google Scholar 

  48. Machanick, P. & Bailey, T. L. MEME-ChIP: motif analysis of large DNA datasets. Bioinformatics 27, 1696–1697 (2011)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  50. Swain, A. et al. Drosophila TDP-43 RNA-binding protein facilitates association of sister chromatid cohesion proteins with genes, enhancers and polycomb response elements. PLoS Genet. 12, e1006331 (2016)

    Article  Google Scholar 

Download references

Acknowledgements

We thank D. Cisneros for help with confocal microscopy, A. Sommer and colleagues at Vienna Biocenter Core Facilities for Illumina sequencing and M. Jaritz for generating the density heat map program. Research in the laboratory of J.-M.P. is supported by Boehringer Ingelheim, the Austrian Science Fund (SFB-F34 and Wittgenstein award Z196-B20) and the Austrian Research Promotion Agency (headquarter grants FFG-834223 and FFG-852936, Laura Bassi Centre for Optimized Structural Studies grant FFG-840283).

Author information

Authors and Affiliations

Authors

Contributions

G.A.B. did most experiments; P.v.d.L. performed ChIP–qPCR and the Nipbl and Wapl ChIP-seq experiments; G.A.B. and R.S. performed bioinformatic analyses of ChIP-seq data; R.S. bioinformatically analysed GRO-seq data; E.A. analysed the RNA-seq data; A.T. generated the conditional Wapl mouse; N.G. provided the conditional Ctcf mouse; G.A.B. and J.-M.P. planned the project, designed experiments and wrote the manuscript.

Corresponding author

Correspondence to Jan-Michael Peters.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks G. Natoli 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 Figure 1 Characterization of conditional Smc3 KO cells.

a, Schematic representation of the WT, floxed (fl) and deleted (∆) Smc3 alleles (after elimination of the neomycin resistance gene). EcoRV fragments, which were used for allele identification by Southern blot analysis with the indicated exon 8 probe, are shown together with their length (in kilobases). b, Southern blot analysis of tail DNA from WT, Smc3fl/+ and Smc3fl/fl mice. c, Absence of Smc3−/−offspring at birth. The genotype of newborn mice from intercrosses of Smc3+/− mice was determined by PCR genotyping. d, Deletion of the floxed Smc3 allele was detected by PCR genotyping in primary Rosa26CreER/+ Smc3fl/+ MEFs at the indicated days after 4-hydroxytamoxifen (OHT) addition. e, The level of Smc3 protein depletion in primary Rosa26CreER/+ Smc3fl/− MEFs was analysed every second day after OHT addition by immunoblot analysis of whole-cell extracts. Control Rosa26CreER/+ Smc3fl/+ MEFs were additionally analysed together with a dilution series of the day 0 sample. A long and a short exposure of the Smc3 immunoblot are shown. f, Proliferation capacity of WT MEFs and Ctcf, Smc3 and Wapl KO MEFs. The indicated serum-starved cells were stimulated with 10% fetal calf serum, and cell numbers were measured every day using a Casy counter. All three KO cells failed to respond to proliferate, in contrast to WT MEFs. g, The efficiency of protein depletion was analysed by immunoblot analysis of chromatin extracts from starved MEFs at day 10 after OHT treatment (Smc3 and Wapl KO cells) or Adeno-Cre virus infection (Ctcf KO cells). The WT chromatin sample was diluted up to 1:32 to estimate the relative reduction in protein levels (CTCF: >4×; Wapl: >8×; Smc3: >16×). Data are shown for one experiment (d, f), one experiment representative of two experiments (e) and one experiment representative of three experiments (g).

Extended Data Figure 2 Cohesin relocation in Ctcf KO MEFs.

Binding of Nipbl, CTCF and cohesin (Stag1 and Scc1) at the Nufip2 (a), Gphn (b) and Ublcp1/Rnf145 (c) genes was determined by ChIP-seq analysis in WT MEFs and Ctcf, Smc3 and Wapl KO MEFs. Data are shown for one experiment representative of two independent experiments (ac; CTCF, Stag1 and Scc1 ChIP-seq) per genotype or one experiment (ac; Nipbl ChIP-seq).

Extended Data Figure 3 Analysis of Ctcf KO-specific cohesin sites.

a, Density profiles of CTCF and Stag1 binding at cohesin sites that are commonly found in WT and Ctcf KO cells. The cohesin sites were subdivided on the basis of the detection or absence of a CTCF peak in Ctcf KO cells as determined by the MACS peak-calling program. b, The enrichment of Stag1 binding (n = 2) relative to input is shown for WT (black) and Ctcf KO cells (purple) at two TSSs, one CTCF site and one TTS. Two independent biological experiments were performed and normalized average values with error bars (s.d.) were plotted. c, d, Cohesin (Stag1 and Scc1) ChIP-seq data of replica (rep) experiments are shown as density heat maps of cohesin binding at the different categories of cohesin-binding sites sites (c) and at active and inactive TSSs (d). Binding data are shown for a region extending from −2.5 kb to +2.5 kb relative to the cohesin peak summit. Heat maps were sorted according to the density of Stag1 binding in Ctcf KO cells (rep 2). A density scale from low (grey) to high (yellow) is shown. As indicated, two or three independent ChIP-seq experiments per genotype were performed. e, Categorization of WT-specific, Ctcf KO-specific and commonly found cohesin sites according to their location at active TSSs (H3K4me2+ H3K9ac+) or non-TSS regions with open chromatin (H3K4me2+ H3K9ac+), poised chromatin (H3K4me2+) and no active chromatin marks (rest). Note that one TSS can bind several cohesin sites. Therefore the number of cohesin-bound TSSs and TSS-bound cohesin sites are not necessarily identical.

Extended Data Figure 4 Cohesin redistribution to transcriptional start sites in Ctcf KO MEFs.

a, Binding of cohesin at TSSs of active and inactive genes, which were defined by RNA-seq analysis. Genes with an RPKM >1 were considered as active, whereas genes with an RPKM <1 were classified as inactive. Pie charts indicate the relative binding of cohesin at all annotated TSSs of the RefSeq genome (mm9) in WT and Ctcf KO MEFs. b, Density heat map of cohesin and Nipbl binding at active and inactive TSSs as defined in a. Active and inactive TSSs were sorted according to the read density of Stag1 binding in Ctcf KO cells (replica 3). c, Binding of cohesin at TSSs of active and inactive genes, which were defined by GRO-seq analysis. Genes with a TPM >1 were considered as active, whereas genes with an TPM <1 were classified as inactive. Pie charts indicate the relative binding of cohesin at all annotated TSSs of the RefSeq genome (mm9) in WT and Ctcf KO cells. d, Density heat map of cohesin and Nipbl binding at active and inactive TSSs as defined in c. Active and inactive TSSs were sorted according to the read density of Stag1 binding in Ctcf KO (replica 3). A density scale from low (grey) to high (yellow) is shown (b, d). e, Heat map of cohesin binding at Nipbl peaks in MEFs of the indicated genotypes. The Nipbl peaks were subdivided according to their TSS localization. Peaks were sorted according to the Stag1 binding density in Ctcf KO cells (replica 3). f, Venn diagram indicating the overlap between Nipbl and cohesin peaks in WT or Ctcf KO MEFs. As indicated in b, d and e, two or three independent ChIP-seq experiments per genotype were performed.

Extended Data Figure 5 Identification of CTCF- and cohesin-regulated genes.

a, Scatter plot of gene expression differences between WT and Ctcf KO or Smc3 KO MEFs, on the basis of two and four independent RNA-seq experiments, respectively. The normalized expression data of individual genes in the two cell types are plotted as coefficient values. Each symbol represents one gene. Genes with an expression difference greater than twofold, an adjusted P value <0.1 and an RPKM value >1 in WT or KO cells are coloured in blue or red, corresponding to down- or upregulated genes in the indicated KO MEFs, respectively. For evaluation of the RNA-seq data, see Methods. b, Overlap between CTCF- and cohesin-regulated genes, shown as a Venn diagram. ce, Expression of selected regulated genes in WT (black), Ctcf KO (purple) and Smc3 KO (blue) MEFs. The expression of genes, which are commonly regulated by CTCF and cohesin (c), by CTCF alone (d) or by cohesin alone (e), is shown as normalized average expression value (RPKM) with error bars (s.d.) based on ten (WT), two (Ctcf KO) or four (Smc3 KO) independent RNA-seq experiments. f, h, Minimal correlation between CTCF-dependent gene regulation and cohesin (f) or CTCF (h) binding at active promoters. Genes that were down- or upregulated in Ctcf KO MEFs are shown as the percentage of all genes present in the three indicated gene groups that were defined by the presence or absence of cohesin binding at active TSSs in WT and/or Ctcf KO cells. g, i, Little correlation between Smc3-dependent gene regulation and cohesin (g) or CTCF (i) binding at active promoters. Genes that were down- or upregulated in Smc3 KO MEFs are shown as the percentage of all genes that were defined by the presence or absence of cohesin binding at active TSSs in WT cells.

Extended Data Figure 6 Genomic localization of cohesin in Wapl KO cells.

a, The cohesin-binding sites detected in WT and Wapl KO cells were subdivided into WT-specific, Wapl KO-specific and common peaks, as indicated by the Venn diagram. For each subgroup, the most significant DNA-binding motif is shown. The different motifs were detected with the E value indicated in brackets. b, Heat maps of Nipbl and cohesin binding for the different subgroups. Cohesin and Nipbl peaks at cohesin-binding sites (vertical axis) are shown for a region extending from −2.5 kb to +2.5 kb relative to the cohesin peak summit (horizontal axis) and were sorted in each subgroup according to the density of Stag1 binding in Wapl KO cells (replica 3). A density scale from low (grey) to high (yellow) is shown. The number of independent ChIP-seq experiments per genotype are indicated. c, Venn diagram of Wapl and cohesin peaks in WT MEFs. The individual subgroups were further overlapped with Nipbl sites. d, Density profiles of Scc1 binding are shown for Wapl only sites (green) and cohesin/Wapl common sites (blue). Note that cohesin is also enriched (even though at a low level, which was not detected by the peak-calling algorithm we used) at ‘Wapl only’ sites, consistent with our observation that the association of Wapl with chromatin depends on cohesin49 (Extended Data Fig. 1f). e, Examples of the distribution of Nipbl, Wapl and cohesin (Stag1 and Scc1) at two genomic regions, one on chromosome 11 and the other on chromosome 1, as determined by ChIP-seq analysis in WT MEFs.

Extended Data Figure 7 Cohesin islands in Ctcf Wapl DKO MEFs.

Binding of CTCF, Nipbl and cohesin (Stag1 and Scc1) at the convergently transcribed genes Mier1 and Slc35d1 (a) and Mllt1 and Dnajc1 (b) was determined by ChIP-seq analysis in WT MEFs, Ctcf, Smc3 and Wapl KO MEFs and Ctcf Wapl DKO MEFs. c, d, Time-course analysis of the appearance of cohesin islands upon Ctcf and Wapl deletion in Adeno-Cre infected MEFs. c, Scc1 accumulation in the 3′-region of the convergently transcribed Usp47 and Dkk3 genes at the indicated days after Adeno-Cre infection. d, Density profiles of Scc1 accumulation at convergently transcribed genes in response to Adeno-Cre infection. Scc1 binding was centred in the middle of the intervening region between gene pairs with a similarly strong transcription activity (TPM > 5). Data are shown for one experiment representative of two or three independent experiments (a, b; CTCF, Stag1 and Scc1 ChIP-seq) per genotype or one experiment (a, b; Nipbl ChIP-seq) and one experiment (c, d).

Extended Data Figure 8 Cohesin islands at isolated genes depend on gene transcription.

a, Similar transcriptional activity of individual genes in WT and Ctcf Wapl DKO MEFs. GRO-seq data are shown for three different gene regions in Ctcffl/fl Waplfl/fl MEFs before and after Adeno-Cre infection. b, Density profiles of Scc1 accumulation in WT and Ctcf Wapl DKO cells at all WT cohesin peaks (28,334) or at overlapping canonical peaks (10,390) identified by peak calling in WT and Ctcf Wapl DKO cells. c, The transcriptional activity determines the amount of cohesin accumulation in the 3′-region of isolated genes lacking a neighbouring downstream gene. Density profiles of Scc1 binding are shown for five groups of genes with decreasing GRO-seq signals (TPM values >9, 5–9, 3–5, 1–3 and <1). d, For better visualization of cohesin islands in Wapl KO cells, we restricted our analysis to genes that were >15 kb in length and highly expressed (TPM > 5). The genes were further filtered on the basis of the presence of a cohesin island in Ctcf Wapl DKO cells and the absence of any intragenic CTCF site (assuming that a CTCF site might prevent proper cohesin pushing along the DNA). Density profiles of Scc1 in WT (black), Wapl KO (green) and Ctcf Wapl DKO (turquoise) cells are shown. e, Examples of cohesin island formation in Wapl KO cells are shown for the A230046K03Rik/Appl2 locus. Data are shown for one GRO-seq experiment (a) per treatment and one representative experiment of two or three ChIP-seq experiments (be) per genotype.

Extended Data Figure 9 Transcriptional changes induced by serum stimulation affect positioning of cohesin islands.

a, c, e, g, i, Binding of Scc1 and mRNA profiling upon serum stimulation (20% fetal bovine serum) for 6 h at five genomic regions. The observed differences in cohesin island positioning correlate with increased or decreased mRNA levels of the respective genes as measured by their RPKM value. a, Capn2: 104.88 (0 h) → 248.32 (6 h); c, Dock5: 4.89 → 14.06; e, Abca1: 11.51 → 2.27; g, Pphln1: 5.13 → 8.00 and Prickle1: 14.19 → 7.86; i, Mast4: 7.70 → 16.68). b, d, f, h, j, Visualization of the altered shape of cohesin islands by plotting the cumulative sum of reads per nucleotide starting from −20 kb to +20 kb after the TTSs of Capn2 (b), Dock5 (d), Abca1 (f), Pphln1 (h) and Mast4 (j) genes (see Methods). Data are shown for one experiment.

Extended Data Figure 10 Transcriptional inhibition and disappearance of cohesin islands.

a, c, Loss of cohesin islands in response to transcriptional inhibition by actinomycin D (Act D; 5 μg ml−1) for 2.5 and 5 h followed by Scc1 profiling in Ctcf Wapl DKO cells. b, d, Disappearance and reappearance of cohesin islands in response to inhibition of RNA polymerase II elongation by DRB (100 μM) and its subsequent removal in Ctcf Wapl DKO cells, respectively. The same convergently transcribed gene pairs are shown for the Act D and DRB experiments (a, b, Arid2 and Scaf11; c, d, Ube2k and Pds5a). Note that Nipbl localization in Drosophila is not affected by transcriptional inhibitors50, implying that Nipbl-cohesin interactions may not be sufficient to explain cohesin accumulation at TSSs in DRB-treated cells. Data are shown for one experiment (ad). e, f, Speculative models of how transcription could move one19 (e) or two18 (f) cohesin rings to mediate loop extrusion.

Supplementary information

Supplementary Information

This file contains: Supplementary Figure 1, the source data for western blots, southern blot and DNA agarose gel; Supplementary Table 1, ChIP-seq experiments used in this study; Supplementary Table 2 RNA-seq and GRO-seq experiments used in this study and Supplementary Table 3, a list of Oligonucleotide primers. Supplementary Figures 1a, d, e are uncropped Western blot exposures for (a) Fig. 1a, (d) Extended Data Fig. 1e and (e) Extended Data Fig. 1g. Supplementary Figure 1b shows uncropped Southern blot exposure for Extended Data Fig. 1b and Supplementary Figure 1c shows uncropped agarose gel for Extended Data Fig. 1d. Supplementary Table 1 contains all ChIP-seq experiments used in this study. The number of peaks identified by MACS peak calling is shown for each ChIP-seq experiment, and the common peaks between replica experiments are indicated. In the “merged replica ChIP” column, the ChIP data of the indicated rows were combined into one dataset and analyzed as described in Online Methods. n.a., not applicable. Supplementary Table 2 contains all RNA-seq and GRO-seq data analyzed in the manuscript. (PDF 503 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Busslinger, G., Stocsits, R., van der Lelij, P. et al. Cohesin is positioned in mammalian genomes by transcription, CTCF and Wapl. Nature 544, 503–507 (2017). https://doi.org/10.1038/nature22063

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature22063

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