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.

Polycomb repressive complex PRC1 spatially constrains the mouse embryonic stem cell genome

Abstract

The Polycomb repressive complexes PRC1 and PRC2 maintain embryonic stem cell (ESC) pluripotency by silencing lineage-specifying developmental regulator genes1. Emerging evidence suggests that Polycomb complexes act through controlling spatial genome organization2,3,4,5,6,7,8,9. We show that PRC1 functions as a master regulator of mouse ESC genome architecture by organizing genes in three-dimensional interaction networks. The strongest spatial network is composed of the four Hox gene clusters and early developmental transcription factor genes, the majority of which contact poised enhancers. Removal of Polycomb repression leads to disruption of promoter-promoter contacts in the Hox gene network. In contrast, promoter-enhancer contacts are maintained in the absence of Polycomb repression, with accompanying widespread acquisition of active chromatin signatures at network enhancers and pronounced transcriptional upregulation of network genes. Thus, PRC1 physically constrains developmental transcription factor genes and their enhancers in a silenced but poised spatial network. We propose that the selective release of genes from this spatial network underlies cell fate specification during early embryonic development.

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: PRC1 organizes three-dimensional promoter-promoter contact networks in mouse ESCs.
Figure 2: Validation of a PRC1-dependent three-dimensional promoter-promoter contact network in mouse ESCs.
Figure 3: PRC1 is a key regulator of three-dimensional genome architecture in mouse ESCs.
Figure 4: PRC1-bound promoters preferentially contact poised enhancers.
Figure 5: The Hox spatial network is poised for transcription.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

References

  1. Simon, J.A. & Kingston, R.E. Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol. Cell 49, 808–824 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bantignies, F. et al. Polycomb-dependent regulatory contacts between distant Hox loci in Drosophila. Cell 144, 214–226 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. 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 

  4. Eskeland, R. et al. Ring1B compacts chromatin structure and represses gene expression independent of histone ubiquitination. Mol. Cell 38, 452–464 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Denholtz, M. et al. Long-range chromatin contacts in embryonic stem cells reveal a role for pluripotency factors and polycomb proteins in genome organization. Cell Stem Cell 13, 602–616 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Isono, K. et al. SAM domain polymerization links subnuclear clustering of PRC1 to gene silencing. Dev. Cell 26, 565–577 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Endoh, M. et al. Histone H2A mono-ubiquitination is a crucial step to mediate PRC1-dependent repression of developmental genes to maintain ES cell identity. PLoS Genet. 8, e1002774 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kondo, T. et al. Polycomb potentiates Meis2 activation in midbrain by mediating interaction of the promoter with a tissue-specific enhancer. Dev. Cell 28, 94–101 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Vieux-Rochas, M., Fabre, P.J., Leleu, M., Duboule, D. & Noordermeer, D. Clustering of mammalian Hox genes with other H3K27me3 targets within an active nuclear domain. Proc. Natl. Acad. Sci. USA 112, 4672–4677 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hughes, J.R. et al. Analysis of hundreds of cis-regulatory landscapes at high resolution in a single, high-throughput experiment. Nat. Genet. 46, 205–212 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Dryden, N.H. et al. Unbiased analysis of potential targets of breast cancer susceptibility loci by Capture Hi-C. Genome Res. 24, 1854–1868 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Mifsud, B. et al. High-resolution capture Hi-C to map long-range promoter contacts in human cells. Nat. Genet. 47, 598–606 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Schoenfelder, S. et al. The pluripotent regulatory circuitry connecting promoters to their long-range interacting elements. Genome Res. 25, 582–597 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Azuara, V. et al. Chromatin signatures of pluripotent cell lines. Nat. Cell Biol. 8, 532–538 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Bernstein, B.E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Tavares, L. et al. RYBP-PRC1 complexes mediate H2A ubiquitylation at polycomb target sites independently of PRC2 and H3K27me3. Cell 148, 664–678 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Montavon, T. et al. A regulatory archipelago controls Hox genes transcription in digits. Cell 147, 1132–1145 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Noordermeer, D. et al. Temporal dynamics and developmental memory of 3D chromatin architecture at Hox gene loci. eLife 3, e02557 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Noordermeer, D. et al. The dynamic architecture of Hox gene clusters. Science 334, 222–225 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Williamson, I. et al. Anterior-posterior differences in HoxD chromatin topology in limb development. Development 139, 3157–3167 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. de Wit, E. et al. The pluripotent genome in three dimensions is shaped around pluripotency factors. Nature 501, 227–231 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Zentner, G.E., Tesar, P.J. & Scacheri, P.C. Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res. 21, 1273–1283 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Endoh, M. et al. Polycomb group proteins Ring1A/B are functionally linked to the core transcriptional regulatory circuitry to maintain ES cell identity. Development 135, 1513–1524 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Blackledge, N.P. et al. Variant PRC1 complex–dependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell 157, 1445–1459 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Rosa, S. et al. Physical clustering of FLC alleles during Polycomb-mediated epigenetic silencing in vernalization. Genes Dev. 27, 1845–1850 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ghavi-Helm, Y. et al. Enhancer loops appear stable during development and are associated with paused polymerase. Nature 512, 96–100 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Zhou, X. et al. Exploring long-range genome interactions using the WashU Epigenome Browser. Nat. Methods 10, 375–376 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Atsuta, T. et al. Production of monoclonal antibodies against mammalian Ring1B proteins. Hybridoma 20, 43–46 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. de Napoles, M. et al. Polycomb group proteins Ring1A/B link ubiquitylation of histone H2A to heritable gene silencing and X inactivation. Dev. Cell 7, 663–676 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Silva, J. et al. Establishment of histone H3 methylation on the inactive X chromosome requires transient recruitment of Eed-Enx1 polycomb group complexes. Dev. Cell 4, 481–495 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Dekker, J., Rippe, K., Dekker, M. & Kleckner, N. Capturing chromosome conformation. Science 295, 1306–1311 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Bolland, D.J., King, M.R., Reik, W., Corcoran, A.E. & Krueger, C. Robust 3D DNA FISH using directly labeled probes. J. Vis. Exp. doi:10.3791/50587 (2013).

  36. Parkhomchuk, D. et al. Transcriptome analysis by strand-specific sequencing of complementary DNA. Nucleic Acids Res. 37, e123 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Levin, J.Z. et al. Comprehensive comparative analysis of strand-specific RNA sequencing methods. Nat. Methods 7, 709–715 (2010).

    Article  CAS  PubMed  PubMed Central  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  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  40. Ross-Innes, C.S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold changes and dispersion for RNA-Seq data with DESeq2. bioRxiv doi:10.1101/002832 (2014).

  43. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Elderkin, Fraser and Luscombe groups for discussions and J. Houseley and P. Rugg-Gunn for commenting on the manuscript. We thank F. Krueger for help with data processing and formatting. We thank R.J. Klose and N. Brockdorff for sequencing. We thank D. Bolland, J. Martins and A. Corcoran for help and advice with the three-dimensional DNA FISH and MetaCyte data analyses. This work was funded by the Wellcome Trust (WT085102MA) (S.E.), the Biotechnology and Biological Science Research Council, the Medical Research Council UK (P.F.) and the European Union Framework Programme 7 Epigenesys Network of Excellence (N.M.L.).

Author information

Authors and Affiliations

Authors

Contributions

S.S. conceptualized, designed and performed promoter CHi-C and helped with data interpretation and writing of the manuscript. R.S. analyzed promoter-promoter (promoter CHi-C) data and performed network analysis. A.D. performed nuclear RNA-seq, analyzed promoter-genome (promoter CHi-C) data, gene expression and ChIP-seq data, and helped write the manuscript. B.-M.J. performed 3C-PCR and commented on the manuscript. H.A. performed three-dimensional DNA FISH, analyzed MetaCyte data and helped with CHi-C data analysis. B.M. analyzed ChIP-seq data, performed promoter-promoter contact enrichment analysis and commented on the manuscript. E.D. performed experiments and histone ChIP-seq. L.M. performed three-dimensional DNA FISH, analyzed MetaCyte data and helped with CHi-C data analysis. F.T.-C. mapped ChIP-seq data and analyzed promoter-genome enrichments. M.F.-M. helped with promoter CHi-C protocol development and wild-type ESC promoter CHi-C. W.J. analyzed ChIP-seq data. A.S.-P. analyzed three-dimensional DNA FISH data. S.W.W. helped with the mapping and analysis of promoter CHi-C data. K.T. carried out sequencing. B.H., E.L., C.S.O., S.A. and S.W.W. designed and provided the capture system. H.K. provided RING1A-knockout ESCs and helped with manuscript preparation. P.F. helped with study design, data interpretation and manuscript preparation. N.M.L. was involved in study design, data interpretation and manuscript preparation. S.E. conceptualized and designed the study, designed and performed experiments, interpreted data and wrote the manuscript.

Corresponding author

Correspondence to Sarah Elderkin.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of Hox network promoters in WT ESCs.

(a) Contact enrichment for promoters occupied by the indicated chromatin proteins, for promoter-promoter contacts >10 Mb apart. (b) Circos plot of significant cis and trans Promoter CHi-C contacts between Hox cluster gene promoters in WT ESCs. Inner circle, chromosomes; outer circle, gene names; edges, significant promoter contacts weighted by read count. (c) Genome-wide virtual 4C promoter-promoter contacts using the Hoxa cluster as a viewpoint, generated from WT ESC Promoter CHi-C data. Significant promoter-promoter contacts >10 Mb apart are marked with arrows. The table indicates the number of raw interactions originating from promoter fragments in each Hox cluster and the number of raw interactions (i) within a Hox cluster, (ii) in cis but outside the cluster or (iii) in trans. (d) Gene Ontology analysis (DAVID) showing biological processes enriched among Hox network genes compared to all genes. (e) Protein domain enrichment analysis (DAVID and PROSITE) among all Hox network genes (left) and Hox cluster–contacting genes (right) compared to all genes. (f) RING1B peak enrichment at promoters. P values were calculated by Wilcoxon rank-sum tests.

Supplementary Figure 2 PRC1 dependency of the Hox spatial network and Hox loci 3D conformations.

(a) Top left, immunoblot for RING1B, NANOG and OCT4 in total cell extracts from WT, RING1A-KO and RING1A/B-dKO ESCs. Top middle, immunoblot for H2AK119ub1 in histone extracts from WT, RING1A-KO and RING1A/B-dKO ESCs and Coomassie Brilliant Blue (CBB). Top right, alkaline phosphatase staining of RING1A-KO and RING1A/B-dKO cells 48 h after treatment with 800 nM tamoxifen. Bottom, immunofluorescence for RING1B (green) and OCT4 (red) in WT, RING1A-KO and RING1A/B-dKO ESCs. (b,c) Hox network Circos contact map in RING1A-KO (b) and RING1A/B-dKO (c) ESCs. Inner circle, chromosomes; outer circle, gene names; red dots, Hox network RING1B-bound promoters; edges, significant promoter contacts (orange, weighted by read count) and non-significant contacts (grey, minimum of three reads). (df) CHi-C promoter-promoter contacts within the Hoxb, Hoxc and Hoxd clusters for WT, RING1A-KO and RING1A/B-dKO ESCs, displayed using the WashU Epigenome Browser30. Upper tracks show genomic location, RefSeq genes and WT RING1B ChIP-seq signal17.

Supplementary Figure 3 Validation of Hox network contacts by double-label 3D DNA FISH.

(a,b) Double-label 3D DNA FISH in WT, RING1A-KO and RING1A/B-dKO ESCs for (a) the Hoxb cluster (green), Lhx1 (red) and Gm11443 (red) and (b) the Hoxd cluster (green), Lmx1b (red) and Gm13481 (red). Upper panels show the genomic locations of the gene loci analyzed by DNA FISH. Middle panels show representative double-label 3D DNA FISH images. Scale bar, 5 µm. Below are the cumulative frequency distribution plots, the percentage of alleles colocalizing at increasing distance cutoffs and the percentage of interprobe distances below the specified cutoff, set at the distance that included the lowest quintile of measurements for Hox network member alleles in WT or RING1A-KO ESCs. P values were calculated by Mann-Whitney test (comparison of interprobe distances within the same cell) and Kruskall-Wallis/Dunn’s multiple-comparisons test (comparison of interprobe distances between RING1A-KO and RING1A/B-dKO cells).

Supplementary Figure 4 Validation of Hoxa, Hoxb and Hoxc contacts by 3C-PCR.

(a) 3C-PCR validation of Hoxa cis contacts, as depicted in Figure 2, using a second biological replicate. (b,c) 3C-PCR validation of Hoxb (b) and Hoxc (c) cis contacts for regions identified by Promoter CHi-C as making PRC1-dependent contacts with Hoxb (b) (Lhx1 and Cbx8/4) and Hoxc (c) (Wnt7b) and control regions identified as non-contacting regions for Hoxb (b) (Hlf and Gna13) and Hoxc (c) (Cntn1). Left, genomic locations of gene loci analyzed by 3C-PCR, RING1B ChIP-seq signal from WT ESCs17 and virtual 4C (v4C) cis contacts using the Hoxb (b) and Hoxc (c) clusters as viewpoints, generated from Promoter CHi-C data from WT, RING1A-KO and RING1A/B-dKO ESCs. Right, 3C-PCR results for selected regions for two independent biological replicates from WT, RING1A-KO and RING1A/B-dKO cells.

Supplementary Figure 5 Validation of Hoxd and Hist1 cluster contacts by 3C-PCR.

(a,b) 3C-PCR validation of Hoxd (a) and Hist1 (b) cluster cis contacts for regions identified by Promoter CHi-C as making PRC1-dependent contacts with Hoxd (a) (Lmx1b and Pax6) and PRC1-independent contacts with Hist1h2ae (b) (Hist1h3e and Hist1h4i) and control regions identified as non-contacting regions for Hoxd (a) (Gtdc1 and Rag1) and Hist1h2ae (b) (Vmn1r198). Top, genomic locations of the gene loci analyzed by 3C-PCR, RING1B ChIP-seq signal from WT ESCs17 and virtual 4C (v4C) cis contacts, using the Hoxd cluster (a) and Hist1h2ae (b) as viewpoints, generated from Promoter CHi-C data from WT, RING1A-KO and RING1A/B-dKO ESCs. Below, 3C-PCR results for selected regions for two independent biological replicates from WT, RING1A-KO and RING1A/B-dKO cells.

Supplementary Figure 6 Polycomb promoter categories and the contribution of PRC2 to Hox network contacts.

(a) Left, Venn diagram showing the number of promoters in the PRC2-only, PRC1/PRC2 and PRC1-only categories. Bars show the percentage of promoters in each category that also overlap an H3K27me3 peak (dark gray), and the actual number of promoters are reported. Right, H3K27me3 peak fold enrichment at PRC2-only, PRC1/PRC2 and PRC1-only promoters (considering only those promoters in each category that overlap an H3K27me3 peak). (b) RING1B (left), SUZ12 (middle) and EZH2 (right) peak fold enrichment at PRC1-only, PRC2-only or PRC1/PRC2-occupied promoters as appropriate. P values were calculated by Wilcoxon rank-sum test. (c) Left, immunoblot for EED, EZH2, OCT4 and NANOG in total cell extracts from Eed+/+ and Eed−/– ESCs. Middle, immunoblot for H3K27me3 in histone extracts from Eed+/+ and Eed−/– ESCs. Right, alkaline phosphatase staining of Eed+/+ and Eed−/– ESCs. (dg) 3C-PCR validation of Hoxb (d), Hoxc (e), Hoxd (f) and Hist1 (g) cis contacts for two independent biological replicates from Eed+/+ and Eed−/– ESCs.

Supplementary Figure 7 Effect of RING1A and RING1B double-knockout on the spatial contacts of PRC1-independent genes.

(a) 3D FISH validation of OCT4/SOX2/NANOG promoter-promoter networks. Left, genomic locations of OSN and control gene loci analyzed by 3D DNA FISH, RING1B ChIP-seq signal from WT ESCs17 and virtual 4C (v4C) contacts, generated from Promoter CHi-C data, using Ranbp10 as a viewpoint. Right top, representative double-label 3D DNA FISH images from WT, RING1A-KO and RING1A/B-dKO cells. Scale bar, 5 µm. Right middle, the cumulative frequency distribution plots show the percentage of alleles colocalizing at increasing distance cutoffs. Lower right, percentage of interprobe distances below the specified cutoff, set at the distance that included the lowest quintile of measurements for OCT4/SOX2/NANOG network member alleles in WT or RING1A-KO ESCs. P values were calculated by Mann-Whitney test (comparison of interprobe distances within the same cell) and Kruskall-Wallis/Dunn’s multiple-comparisons test (comparison of interprobe distances between RING1A-KO and RING1A/B-dKO cells). (b) Contact enrichment for non-PRC1/PRC2 promoters that are adjacent to PRC1/PRC2-bound promoters and an equal number of randomly selected non-PRC1/PRC2 promoters.

Supplementary Figure 8 RING1B promoter-enhancer contacts and enhancer status in RING1A-KO and RING1A/B-dKO cells.

(a) Stacked bar chart displaying the average number of significant enhancer contacts per promoter for poised, active and intermediate enhancers (categories as depicted in Fig. 4a). (b) Proportion of promoters contacting one or more intermediate or active enhancers in WT ESCs (gray) and maintaining one or more poised enhancer contacts in RING1A-KO (blue) and RING1A/B-dKO (green) cells. P values were calculated by Fisher’s exact test (WT ESC proportions). (c) Expression status of promoters that contact PRC1/PRC2-bound promoters in WT ESCs (left) and that form new contacts with PRC1/PRC2-bound promoters in RING1A/B-dKO cells (right). (d) Proportion of promoters that gain contacts with active enhancers in RING1A/B-dKO cells. (e) Gene expression changes in RING1A/B-dKO cells relative to RING1A-KO cells for RING1B-bound promoters that gain contacts with active enhancers. P values were calculated by Wilcoxon rank-sum test. (f) Heat map of input-normalized log2 (RPKM) values in RING1A-KO and RING1A/B-dKO cells for H3K4me1, H3K27me3 and H3K27ac at all enhancers defined in WT ESCs. Each line in the heat map represents an individual enhancer, grouped by WT enhancer status and ordered within each group by H3K4me1 values in RING1A-KO cells. The poised enhancer section of the heat map is shown at higher magnification. (g) Changes in H3K4me1, H3K27me3 and H3K27ac occupancy at intermediate and active enhancers that maintain contacts with RING1B-bound promoters in RING1A/B-dKO cells. (h) Changes in H3K27ac at intermediate enhancers that maintain contacts with RING1B-occupied non-Hox network or Hox network promoters in RING1A/B-dKO cells.

References for Supplementary Material:

44. Shen, Y. et al. A map of the cis-regulatory sequences in the mouse genome. Nature 488, 116–120 (2012).

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

46. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

47. Kaneko, S., Son, J., Shen, S. S., Reinberg, D. & Bonasio, R. PRC2 binds active promoters and contacts nascent RNAs in embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1258–1264 (2013).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8. (PDF 2281 kb)

Supplementary Table 1

Promoter fragment baits in different categories. (XLSX 1374 kb)

Supplementary Table 2

Next-generation sequencing statistics for promoter CHi-C, nuclear RNA-seq and ChIP-seq. (XLSX 17 kb)

Supplementary Table 3

Publically available data sets used. (XLSX 10 kb)

Supplementary Table 4

Enhancer fragment baits in different enhancer classes. (XLSX 6818 kb)

Supplementary Table 5

BACs and primer sequences used for 3C-PCR and 3D DNA FISH. (XLSX 11 kb)

Supplementary Table 6

Interprobe distances for 3D DNA FISH. (XLSX 168 kb)

Supplementary Data Set

Full-length gels for 3C analysis in the main figures. (PDF 46791 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schoenfelder, S., Sugar, R., Dimond, A. et al. Polycomb repressive complex PRC1 spatially constrains the mouse embryonic stem cell genome. Nat Genet 47, 1179–1186 (2015). https://doi.org/10.1038/ng.3393

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3393

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