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:

Cohesin promotes stochastic domain intermingling to ensure proper regulation of boundary-proximal genes

Abstract

The human genome can be segmented into topologically associating domains (TADs), which have been proposed to spatially sequester genes and regulatory elements through chromatin looping. Interactions between TADs have also been suggested, presumably because of variable boundary positions across individual cells. However, the nature, extent and consequence of these dynamic boundaries remain unclear. Here, we combine high-resolution imaging with Oligopaint technology to quantify the interaction frequencies across both weak and strong boundaries. We find that chromatin intermingling across population-defined boundaries is widespread but that the extent of permissibility is locus-specific. Cohesin depletion, which abolishes domain formation at the population level, does not induce ectopic interactions but instead reduces interactions across all boundaries tested. In contrast, WAPL or CTCF depletion increases inter-domain contacts in a cohesin-dependent manner. Reduced chromatin intermingling due to cohesin loss affects the topology and transcriptional bursting frequencies of genes near boundaries. We propose that cohesin occasionally bypasses boundaries to promote incorporation of boundary-proximal genes into neighboring domains.

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: Boundary permissibility is a widespread feature of the human genome.
Fig. 2: Variable interactions across boundaries occur independently from intra-domain compaction.
Fig. 3: Cohesin promotes interactions within and across domain boundaries.
Fig. 4: WAPL and CTCF restrict cohesin-dependent interactions across domain boundaries.
Fig. 5: Cohesin alters the topological context of boundary-proximal genes.
Fig. 6: Cohesin alters the transcriptional bursting frequency of boundary-proximal genes.

Similar content being viewed by others

Data availability

All data that support the findings of this study are included in manuscript or are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Bickmore, W. A. The spatial organization of the human genome. Annu. Rev. Genomics Hum. Genet. 14, 67–84 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Gibcus, J. H. & Dekker, J. The hierarchy of the 3D genome. Mol. Cell 49, 773–782 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Cremer, T. & Cremer, M. Chromosome territories. Cold Spring Harb. Perspect. Biol. 2, a003889 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Hansen, A. S., Pustova, I., Cattoglio, C., Tjian, R. & Darzacq, X. CTCF and cohesin regulate chromatin loop stability with distinct dynamics. eLife 6, e257762017.

  5. Kim, Y. H. et al. Rev-erbα dynamically modulates chromatin looping to control circadian gene transcription. Science 359, 1274–1277 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. 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  PubMed  PubMed Central  Google Scholar 

  8. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320.e24 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhan, Y. et al. Reciprocal insulation analysis of Hi-C data shows that TADs represent a functionally but not structurally privileged scale in the hierarchical folding of chromosomes. Genome Res. 27, 479–490 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sexton, T. et al. Three-dimensional folding and functional organization principles of the Drosophila genome. Cell 148, 458–472 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Le Dily, F. et al. Distinct structural transitions of chromatin topological domains correlate with coordinated hormone-induced gene regulation. Genes Dev. 28, 2151–2162 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Sun, F. et al. Promoter-enhancer communication occurs primarily within insulated neighborhoods. Mol. Cell 73, 250–263.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Dowen, J. M. et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159, 374–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lupianez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161, 1012–1025 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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  PubMed  PubMed Central  Google Scholar 

  18. Goloborodko, A., Marko, J. F. & Mirny, L. A. Chromosome compaction by active loop extrusion. Biophys. J. 110, 2162–2168 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mirny, L. A., Imakaev, M. & Abdennur, N. Two major mechanisms of chromosome organization. Curr. Opin. Cell Biol. 58, 142–152 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  21. Splinter, E. et al. CTCF mediates long-range chromatin looping and local histone modification in the β-globin locus. Genes Dev. 20, 2349–2354 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nora, E. P. et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169, 930–944.e22 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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 

  25. 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  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Bintu, B. et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Finn, E. H. et al. Extensive heterogeneity and intrinsic variation in spatial genome organization. Cell 176, 1502–1515.e10 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Beliveau, B. J. et al. Single-molecule super-resolution imaging of chromosomes and in situ haplotype visualization using Oligopaint FISH probes. Nat. Commun. 6, 7147 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Beliveau, B. J. et al. Versatile design and synthesis platform for visualizing genomes with Oligopaint FISH probes. Proc. Natl Acad. Sci. USA 109, 21301–21306 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Beliveau, B. J. et al. OligoMiner provides a rapid, flexible environment for the design of genome-scale oligonucleotide in situ hybridization probes. Proc. Natl Acad. Sci. USA 115, E2183–E2192 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Beliveau, B. J., Apostolopoulos, N. & Wu, C. T. Visualizing genomes with Oligopaint FISH probes. Curr. Protoc. Mol. Biol. 105, 14.23.1–14.23.20 (2014).

    Google Scholar 

  34. Beliveau, B. J. et al. In situ super-resolution imaging of genomic DNA with OligoSTORM and OligoDNA-PAINT. Methods Mol. Biol. 1663, 231–252 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shin, H. et al. TopDom: an efficient and deterministic method for identifying topological domains in genomes. Nucleic Acids Res. 44, e70 (2016).

    Article  PubMed  CAS  Google Scholar 

  36. ENCODE Project Consortium An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  37. Ollion, J., Cochennec, J., Loll, F., Escude, C. & Boudier, T. TANGO: a generic tool for high-throughput 3D image analysis for studying nuclear organization. Bioinformatics 29, 1840–1841 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chang, L.-H., Ghosh, S. & Noordermeer, D. TADs and their borders: free movement or building a wall?. J. Mol. Biol. 432, 643–652 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Natsume, T., Kiyomitsu, T., Saga, Y. & Kanemaki, M. T. Rapid protein depletion in human cells by auxin-inducible degron tagging with short homology donors. Cell Rep. 15, 210–218 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  41. Eisenberg, E. & Levanon, E. Y. Human housekeeping genes, revisited. Trends Genet. 29, 569–574 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Dorsett, D. & Krantz, I. D. On the molecular etiology of Cornelia de Lange syndrome. Ann. N. Y. Acad. Sci. 1151, 22–37 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, J. et al. Transcriptional dysregulation in NIPBL and cohesin mutant human cells. PLoS Biol. 7, e1000119 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Nir, G. et al. Walking along chromosomes with super-resolution imaging, contact maps, and integrative modeling. PLoS Genet. 14, e1007872 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Cardozo Gizzi, A. M. et al. Microscopy-based chromosome conformation capture enables simultaneous visualization of genome organization and transcription in intact organisms. Mol. Cell 74, 212–222.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Nuebler, J., Fudenberg, G., Imakaev, M., Abdennur, N. & Mirny, L. A. Chromatin organization by an interplay of loop extrusion and compartmental segregation. Proc. Natl Acad. Sci. USA 115, E6697–E6706 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Haarhuis, J. H. I. et al. The cohesin release factor WAPL restricts chromatin loop extension. Cell 169, 693–707.e14 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Thiecke, M. J. et al. Cohesin-dependent and independent mechanisms support chromosomal contacts between promoters and enhancers. Preprint at bioRxiv https://doi.org/10.1101/2020.02.10.941989 (2020).

  49. Bartman, C. R., Hsu, S. C., Hsiung, C. C., Raj, A. & Blobel, G. A. Enhancer regulation of transcriptional bursting parameters revealed by forced chromatin looping. Mol Cell 62, 237–247 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3, 95–98 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Moffitt, J. R. & Zhuang, X. RNA imaging with multiplexed error-robust fluorescence in situ hybridization (MERFISH). Methods Enzymol. 572, 1–49 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rosin, L. F., Nguyen, S. C. & Joyce, E. F. Condensin II drives large-scale folding and spatial partitioning of interphase chromosomes in Drosophila nuclei. PLoS Genet. 14, e1007393 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Ollion, J., Cochennec, J., Loll, F., Escude, C. & Boudier, T. Analysis of nuclear organization with TANGO, software for high-throughput quantitative analysis of 3D fluorescence microscopy images. Methods Mol. Biol. 1228, 203–222 (2015).

    Article  CAS  PubMed  Google Scholar 

  54. Kishi, J. Y. et al. SABER amplifies FISH: enhanced multiplexed imaging of RNA and DNA in cells and tissues. Nat. Methods 16, 533–544 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z. & Zhang, B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 47, W199–W205 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Durand, N. C. et al. Juicebox provides a visualization system for Hi-C contact maps with unlimited zoom. Cell Syst. 3, 99–101 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hnisz, D., Day, D. S. & Young, R. A. Insulated neighborhoods: structural and functional units of mammalian gene control. Cell 167, 1188–1200 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank G. Blobel, L. Rosin, M. Lakadamyali, and members of the Joyce laboratory for helpful discussions and critical reading of the manuscript. In addition, we thank M. Kanemaki for the HCT-116-RAD21-AID cell line and S. Rao and E. Lieberman-Aiden for sharing critical primary datasets. We also thank A. Stout at the Penn Cell and Developmental Biology Microscopy core and C. Eberling from Bruker for assistance with super-resolution imaging and analysis. This work was supported by a Charles E. Kaufman grant from The Pittsburgh Foundation (KA2017-91787) to E.F.J. and NIH grants R35GM128903 to E.F.J. and T32GM008216 to J.M.L.

Author information

Authors and Affiliations

Authors

Contributions

J.M.L. and E.F.J. designed the study. J.M.L., D.S.P., S.C.N. and E.F.J. analyzed the results. J.M.L. and E.F.J. wrote the manuscript. J.M.L. and D.S.P. performed all of the experiments, except the RNA FISH experiments, which were performed by S.C.N. Y.L. performed the Hi-C data analysis. Z.X. and R.Y. generated all of the Oligopaint probes used in this study. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Eric F. Joyce.

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.

Extended data

Extended Data Fig. 1 Genomic landscapes at Oligopaint target regions.

Genomic profiles of loci imaged by FISH. Hi-C contact matrices visualized by Juicebox (v1.9.0)56. Data from HCT-116-RAD21-AID8 cells. Solid and dashed lines indicate domains and subdomains, respectively. The gene density, eigenvectors, and insulation score (computed by the TopDom) are noted below. Insulation score computed prior to (solid) and following 6 hours of auxin treatment (dashed). Published ChIP–seq tracks8 depict protein binding and histone modifications in the HCT-116-RAD21-AID cell line prior to and following 6 hours of auxin treatment (-/+ Auxin). Genomic tracks visualized using Integrative Genomics Viewer57.

Extended Data Fig. 2 Additional information related to Fig. 1.

a, HCT-116-RAD21-AID cells were synchronized at the G1/S transition. Immunofluorescence for CENPF (green) to indicate cells in G2 and PCNA (gray) to mark cells in S phase. DNA (Hoescht stain) is shown in gray in first column. Dashed lines represent nuclear edges. Scale bar equals 5 μm. b, Cumulative frequency distribution of spatial overlap between neighboring domains on chr12:11.6Mb-13.6 Mb (n = 716 chromosomes) and chr22:33.2Mb-36.8 Mb (n = 1410 in asynchronous HCT-116 cells. Overlap normalized to the volume of the upstream domain. n > 716 chromosomes. c, Hi-C contact matrix and Oligopaint designs corresponding to (c-e). d, Representative FISH images of each subdomain and the downstream D2. Scale bar equals 1 μm. Corresponding 3D segmentation of FISH signals below each image. e, Cumulative distribution plot of spatial overlap between the subdomains [S1 (n = 1932); S2 (n = 2283); S3 (n = 1977)] and D2, normalized to the volume of D2. *** P < 0.001, two-tailed Mann-Whitney test. f, Frequency of contact between each subdomain and D2 from data in e. Contact defined as > 500 nm3 overlap. **** P < 0.0001, two-tailed Fisher’s exact test. g, Scatterplot of spatial overlap volume across the strong and weak boundaries on the same allele at the chr22:33.2-36.8 Mb locus. n = 1060 chromosomes. See Fig. 2e for corresponding Oligopaint design.

Extended Data Fig. 3 Boundary permissibility is a widespread feature of the human genome (additional loci).

a, Distribution of spatial overlap between neighboring domains (D1 & D2) at chr1:36.5-40.3 Mb. Overlap normalized to the volume of the upstream domain. n = 2294 chromosomes. b, Distribution of spatial overlap between neighboring domains (D2 & D3) at chr1:36.5-40.3 Mb. Overlap normalized to the volume of the upstream domain. n = 2553 chromosomes. c, Distribution of spatial overlap between neighboring domains (D3 & D4) at chr1:36.5-40.3 Mb. Overlap normalized to the volume of the upstream domain. n = 2502 chromosomes. d, Distribution of spatial overlap between neighboring domains (D4 & D5) at chr1:36.5-40.3 Mb. Overlap normalized to the volume of the upstream domain. n = 9443 chromosomes. e, Distribution of spatial overlap between neighboring domains (D1 & D2) at chr2:217.45-223 Mb. Overlap normalized to the volume of the upstream domain. n = 1850 chromosomes. f, Distribution of spatial overlap between neighboring domains (D2 & D3) at chr2:217.45-223 Mb. Overlap normalized to the volume of the upstream domain. n = 1903 chromosomes. g, Distribution of spatial overlap between neighboring domains (D2 & D3) at chr3:44.2-47.55 Mb. Overlap normalized to the volume of the upstream domain. n = 1657 chromosomes. h, Distribution of spatial overlap between neighboring domains (D2 & D3) at chr12:11.6-13.6 Mb. Overlap normalized to the volume of the upstream domain. n = 1606 chromosomes. i, Distribution of spatial overlap between neighboring domains (S1 & S2) at chr12:11.6-13.6 Mb. Overlap normalized to the volume of the upstream domain. n = 1479 chromosomes. j, Distribution of spatial overlap between neighboring domains (S2 & S3) at chr12:11.6-13.6 Mb. Overlap normalized to the volume of the upstream domain. n = 1912 chromosomes. k, Distribution of spatial overlap between neighboring domains (D1 & D2) at chr19:17.35-18.6 Mb. Overlap normalized to the volume of the upstream domain. n = 1406 chromosomes. l, Distribution of spatial overlap between neighboring domains (S1 & S2) at chr22:33.2-36.8 Mb. Overlap normalized to the volume of the upstream domain. n = 1634 chromosomes. m, Distribution of spatial overlap between neighboring domains (S2 & S3) at chr22:33.2-36.8 Mb. Overlap normalized to the volume of the upstream domain. n = 1640 chromosomes. n, Distribution of spatial overlap between neighboring domains (S4 & S5) at chr22:33.2-36.8 Mb. Overlap normalized to the volume of the upstream domain. n = 1494 chromosomes.

Extended Data Fig. 4 Additional information related to Fig. 2.

a, Localization density per domain quantified from 3D-STORM images. Chr12.D1 (median = 0.0002707, n = 91); Chr12.D2 (median = 0.0002076, n = 91); Chr22.D1 (median = 0.0002376, n = 95); Chr22.D2 (median = 0.0002245, n = 95). b, Scatterplot depicting the relationship between domain volumes on the same chromosome by 3D-STORM on chr12:11.6Mb-13.6 Mb. n = 91 chromosomes. c, Scatterplot depicting the relationship between domain volumes between homologs by 3D-STORM on chr12:11.6Mb-13.6 Mb. n = 82 chromosomes. d, Scatterplot depicting the relationship between domain volumes on the same chromosome by 3D-STORM on chr22:33.2Mb-36.8 Mb. n = 95 chromosomes. e, Scatterplot depicting the relationship between domain volumes between homologs by 3D-STORM on chr22:33.2Mb-36.8 Mb. n = 86 chromosomes. f, Scatterplot of overlap volume (x-axis) versus domain density (y-axis) by 3D-STORM for the chr22:33.2Mb-36.8 Mb locus. n = 95 chromosomes.

Extended Data Fig. 5 Confirmation and quantification of RAD21 degradation.

a, Immunofluorescence for RAD21 (cyan). DNA (Hoescht stain) is shown in gray. Dashed lines represent nuclear edges. Scale bar equals 5 μm. b, Western blot to RAD21 protein in the chromatin-bound fraction of HCT-116-RAD21-AID cells with no auxin treatment (-) or following 6 hours of auxin treatment (+). Histone H3 as loading control. Protein was labeled using fluorescent secondary antibodies. c, Fluorescence quantification of RAD21 and H3 isolated from the chromatin-bound fraction of protein corresponding to blot in b using Image Lab v5.2.1. Protein intensity normalized to total protein per well (via stain-free technology) and presented as fraction of protein observed in untreated (- auxin) conditions; we observe a 96% reduction in chromatin-bound RAD21 following auxin treatment.

Source data

Extended Data Fig. 6 Cohesin promotes interactions across domain boundaries (additional loci).

Hi-C contact matrix and corresponding cumulative distribution of spatial overlap between the neighboring domains prior to or after auxin treatment. Overlap normalized to the volume of the upstream domain. a, Map of chr1:36.5-40.3 Mb for b-e. b, D1 & D2 (-aux n = 2294, +aux n = 2175). c, D2 & D3 (-aux n = 2553, +aux n = 2353). d, D3 & D4 (-aux n = 2502, +aux n = 2488), e. D4 & D5 (-aux n = 9443, +aux n = 8695). f, Map of chr2:217.45-223 Mb for g-h. g, D1 & D2 (-aux n = 1850, +aux n = 1665). h, D2 & D3 (-aux n = 1903, +aux n = 1717). i, Map of chr3:44.2-47.55 Mb for j-k. j, D1 & D2 (-aux n = 1643, +aux n = 2186). k, D2 & D3 (-aux n = 1657, +aux n = 2181). l, Map of chr12:11.6-13.6 Mb for m-o. m, D2 & D3 (-aux n = 1606, +aux n = 1570). n, S1 & S2 (-aux n = 1479, +aux n = 1220). o, S2 & S3 (-aux n = 731, +aux n = 678). p, Map of chr19:17.35-18.6 Mb for q. q, D1 & D2 (-aux n = 1406, +aux n = 1419). r, Map of chr22:33.2-36.8 Mb for s-u. s, S1 & S2 (-aux n = 1634, +aux n = 1703). t, S2 & S3 (-aux n = 1640, +aux n = 1702). u, S4 & S5 (-aux n = 1494, +aux n = 1718). P < 0.001, two-tailed Mann-Whitney test for all domain pairs.

Extended Data Fig. 7 Additional information related to Fig. 3.

a, Minimum distances between localizations contained within each domain on chr12:11.6-13.6 Mb as measured from 3D-STORM data (prior to auxin: median = 0.008292 μm, n = 91; after auxin: median = 0.01234 μm, n = 76). P < 0.001, two-tailed Mann-Whitney test. b, Minimum distances between localizations contained within each domain on chr22:33.2Mb-36.8 Mb as measured from 3D-STORM data (prior to auxin: median = 0.009470 μm, n = 95; after auxin: median = 0.01563 μm, n = 105). P < 0.001, two-tailed Mann-Whitney test. c, Violin plots of domain volumes as measured from 3D-STORM data prior to [Chr12.D1 (median = 0.2629 μm3, n = 91); Chr12.D2 (median = 0.1830 μm3, n = 91); Chr22.D1 (median = 0.2749 μm3, n = 95); Chr22.D2 (median = 0.2320 μm3, n = 95)] or after auxin treatment [Chr12.D1 (median = 0.1703 μm3, n = 91); Chr12.D2 (median = 0.1671 μm3, n = 76); Chr22.D1 (median = 0.2284 μm3, n = 105); Chr22.D2 (median = 0.2213μm3, n = 105)]. Chr12.D1 P = 0.008; Chr12.D2 P = 0.368; Chr22.D1 P = 0.076, Chr22.D2 P = 0.907; two-tailed Mann-Whitney test. d, Violin plots of domain densities as measured from 3D-STORM data prior to [Chr12.D1 (median = 0.0002707, n = 91); Chr12.D2 (median = 0.0002076, n = 91); Chr22.D1 (median = 0.0002376, n = 95); Chr22.D2 (median = 0.0002245, n = 95)] or after auxin treatment [Chr12.D1 (median = 0.0003080, n = 76); Chr12.D2 (median = 0.0002385, n = 76); Chr22.D1 (median = 0.0002651, n = 105); Chr22.D2 (median = 0.0002353, n = 105)]. Chr12.D1 P = 0.247; Chr12.D2 P = 0.014; Chr22.D1 P = 0.025, Chr22.D2 P = 0.602; two-tailed Mann-Whitney test.

Extended Data Fig. 8 Additional information related to Fig. 4.

a, Western blot of NIPBL, WAPL, and CTCF protein of HCT-116 cells following RNAi. H3 as loading control. b, Representative FISH images of neighboring domains on chr12:11.6Mb-13.6 Mb in RNAi control, NIPBL-, WAPL-, or CTCF-depleted cells. Dashed lines represent nuclear edges. Scale bar equals 5 μm (left) or 1 μm (zoomed images, below). c, Spatial overlap between neighboring domains on chr12:11.6Mb-13.6 Mb in control (n = 643), NIPBL- (n = 636), WAPL- (n = 819) or both NIPBL and WAPL- (n = 922) depleted cells. ***P < 0.001, two-tailed Mann-Whitney test. d, Spatial overlap between neighboring domains on chr22:33.2Mb-36.8 Mb in control (n = 1722), NIPBL- (n = 1440), WAPL- (n = 1769) or both NIPBL and WAPL- (n = 1762) depleted cells. Overlap normalized to the volume of the upstream domain. ***P < 0.001, two-tailed Mann-Whitney test. e, Western blot to RAD21 and CTCF in whole cell lysate of HCT-116 cells following RNAi to CTCF and/or 6 hours auxin to degrade RAD21. Alpha tubulin as loading control. Spatial overlap between neighboring domains across six loci (f-k). f, D4 and D5 on chr1:36.5-40.3 Mb in control (n = 2362) or CTCF- (n = 1611) depleted cells. ***P < 0.001. two-tailed Mann-Whitney test. g, D1 and D2 on chr2:217.45-223 Mb in control (n = 1484) or CTCF- (n = 1466) depleted cells. P = 0.604 (ns). two-tailed Mann-Whitney test. h, D2 and D3 on chr2:217.45-223 Mb in control or CTCF-depleted cells prior to (control n = 1145; CTCF n = 1133) or after auxin treatment (control n = 1039; CTCF n = 877). P < 0.001 (***), P = 0.008 (**). two-tailed Mann-Whitney test. i, D1 and D2 on chr3:44.2-47.55 Mb in control or CTCF-depleted cells prior to (control n = 1272; CTCF n = 1215) or after auxin treatment (control n = 1254; CTCF n = 1090). P < 0.001 (***) or P = 0.996. two-tailed Mann-Whitney test. j, D1 and D2 on chr12:11.6Mb-13.6 Mb in control or CTCF-depleted cells prior to (control n = 1236; CTCF n = 1250) or after auxin treatment (control n = 1174; CTCF n = 1231). P < 0.001 (***) or P = 0.314. two-tailed Mann-Whitney test. k, D1 and D2 on chr22:33.2-36.8 Mb in control or CTCF-depleted cells prior to (control n = 1258; CTCF n = 1370) or after auxin treatment (control n = 1170; CTCF n = 1157). P < 0.001 (***) or P = 0.89, two-tailed Mann-Whitney test.

Source data

Extended Data Fig. 9 Additional information related to Fig. 5.

a, Hi-C contact matrix chr12:11.6-13.6 Mb and corresponding Oligopaint design for (b-c). Blue represents the boundary proximal gene, CREBL2. b, Cartoon representations for three possible interactions between boundary-proximal genes and their neighboring domains: domain maintenance, switching, and exclusion (top). Representative images of three-color FISH to the chr12:11.6-13.6 Mb locus illustrating the three domain configurations (middle); scale bar equals 1 μm. Corresponding 3D segmentation of FISH signals below each image. c, Frequencies of domain configurations at chr12:11.6-13.6 Mb prior to (n = 1074) or after auxin treatment (n = 979). P < 0.0001 (***) or P = 0.0046 (**), two-sided Fisher’s exact test.

Extended Data Fig 10 Additional information related to Fig. 6.

a, Distribution of domain sizes that harbor either a significantly differentially expressed (median = 183734, n = 4196) or non-differentially expressed genes (median = 194447, n = 8026) in the HCT-116 cell line following auxin treatment. P = 0.102, two-tailed Mann-Whitney test. b, Scatterplot of log2(fold change)] of HCT-116 differentially expressed genes (DEGs) versus the distance between their TSS and the center of the nearest domain boundary. c, Distance to nearest super enhancer defined by H3K27ac signal58 in HCT-116 cells for DEGs with >30% fold change in expression following auxin treatment. Genes were categorized by their proximity to a domain boundary (< 5 kb or > 5 kb away) and whether they were up or down regulated following auxin treatment. d, Gene ontology enrichment analysis for HCT-116 differentially expressed genes (> 30% fold change, n = 68) within 5 kb of a domain boundary. e, Gene ontology enrichment analysis for HCT-116 differentially expressed genes (> 30% fold change, n = 1593) not within 5 kb of a domain boundary. f, Scatterplot of [fold change] of differentially expressed genes (DEGs) associated with CdLS versus the distance between their TSS and the center of the nearest domain boundary in GM12878 cells. n = 1569, P = 0.0017, Spearman correlation. g, Fraction of genes that are up or down regulated in CdLS at binned distances from the nearest domain boundary. h, Fold change in burst volume by RNA FISH in HCT-116 cells following auxin treatment. Each dot represents the fold change in average burst volume per biological replicate; horizonal line indicates mean.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Reporting Summary

Source data

Source Data Extended Data Fig. 5

Unprocessed full Western Blots corresponding to Extended Data Fig. 5.

Source Data Extended Data Fig. 8

Unprocessed full Western Blots corresponding to Extended Data Fig. 8.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luppino, J.M., Park, D.S., Nguyen, S.C. et al. Cohesin promotes stochastic domain intermingling to ensure proper regulation of boundary-proximal genes. Nat Genet 52, 840–848 (2020). https://doi.org/10.1038/s41588-020-0647-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-020-0647-9

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