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:

Smchd1 regulates long-range chromatin interactions on the inactive X chromosome and at Hox clusters

Abstract

The regulation of higher-order chromatin structure is complex and dynamic, and a full understanding of the suite of mechanisms governing this architecture is lacking. Here, we reveal the noncanonical SMC protein Smchd1 to be a novel regulator of long-range chromatin interactions in mice, and we add Smchd1 to the canon of epigenetic proteins required for Hox-gene regulation. The effect of losing Smchd1-dependent chromatin interactions has varying outcomes that depend on chromatin context. At autosomal targets transcriptionally sensitive to Smchd1 deletion, we found increased short-range interactions and ectopic enhancer activation. In contrast, the inactive X chromosome was transcriptionally refractive to Smchd1 ablation, despite chromosome-wide increases in short-range interactions. In the inactive X, we observed spreading of trimethylated histone H3 K27 (H3K27me3) domains into regions not normally decorated by this mark. Together, these data suggest that Smchd1 is able to insulate chromatin, thereby limiting access to other chromatin-modifying proteins.

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: Smchd1 is required to maintain higher-order chromatin organization at target loci and to maintain gene silencing at a subset of its autosomal targets.
Fig. 2: Smchd1 deletion results in activation of autosomal enhancers and increased promoter-enhancer interactions.
Fig. 3: Smchd1 is required to maintain long-range interactions between the HoxB cluster and linked clustered gene families.
Fig. 4: Smchd1 is required for developmentally regulated Hox-gene silencing in vitro and in vivo.
Fig. 5: Smchd1 deletion appears to alter the conformation of the Xi but is not required to maintain gene silencing on the Xi.
Fig. 6: Smchd1 limits H3K27me3 spreading on the Xi, though it is not required to maintain Xi compaction.
Fig. 7: An insulating model for Smchd1 function.

Similar content being viewed by others

References

  1. Bouwman, B. A. & de Laat, W. Getting the genome in shape: the formation of loops, domains and compartments. Genome Biol. 16, 154 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Kakui, Y. & Uhlmann, F. SMC complexes orchestrate the mitotic chromatin interaction landscape. Curr. Genet. 64, 335–339 (2018).

    Article  PubMed  CAS  Google Scholar 

  4. 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 

  5. 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  PubMed  PubMed Central  CAS  Google Scholar 

  6. Schmitt, A. D., Hu, M. & Ren, B. Genome-wide mapping and analysis of chromosome architecture. Nat. Rev. Mol. Cell Biol. 17, 743–755 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. 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 

  8. 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  PubMed  PubMed Central  CAS  Google Scholar 

  9. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Kundu, S. et al. Polycomb repressive complex 1 generates discrete compacted domains that change during differentiation. Mol. Cell 65, 432–446.e5 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Blewitt, M. E. et al. SmcHD1, containing a structural-maintenance-of-chromosomes hinge domain, has a critical role in X inactivation. Nat. Genet. 40, 663–669 (2008).

    Article  PubMed  CAS  Google Scholar 

  13. Chiu, A., Revenkova, E. & Jessberger, R. DNA interaction and dimerization of eukaryotic SMC hinge domains. J. Biol. Chem. 279, 26233–26242 (2004).

    Article  PubMed  CAS  Google Scholar 

  14. Haering, C. H., Löwe, J., Hochwagen, A. & Nasmyth, K. Molecular architecture of SMC proteins and the yeast cohesin complex. Mol. Cell 9, 773–788 (2002).

    Article  PubMed  CAS  Google Scholar 

  15. Hirano, M. & Hirano, T. Hinge-mediated dimerization of SMC protein is essential for its dynamic interaction with DNA. EMBO J. 21, 5733–5744 (2002).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Chen, K., Czabotar, P. E., Blewitt, M. E. & Murphy, J. M. The hinge domain of the epigenetic repressor Smchd1 adopts an unconventional homodimeric configuration. Biochem. J. 473, 733–742 (2016).

    Article  PubMed  CAS  Google Scholar 

  17. Chen, K. et al. Genome-wide binding and mechanistic analyses of Smchd1-mediated epigenetic regulation. Proc. Natl Acad. Sci. USA 112, E3535–E3544 (2015).

    Article  PubMed  CAS  Google Scholar 

  18. Brideau, N. J. et al. Independent mechanisms target SMCHD1 to trimethylated histone H3 lysine 9-modified chromatin and the inactive X chromosome. Mol. Cell. Biol. 35, 4053–4068 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Chen, K. et al. The epigenetic regulator Smchd1 contains a functional GHKL-type ATPase domain. Biochem. J. 473, 1733–1744 (2016).

    Article  PubMed  CAS  Google Scholar 

  20. Blewitt, M. E. et al. An N-ethyl-N-nitrosourea screen for genes involved in variegation in the mouse. Proc. Natl Acad. Sci. USA 102, 7629–7634 (2005).

    Article  PubMed  CAS  Google Scholar 

  21. Lemmers, R. J. L. F. et al. Digenic inheritance of an SMCHD1 mutation and an FSHD-permissive D4Z4 allele causes facioscapulohumeral muscular dystrophy type 2. Nat. Genet. 44, 1370–1374 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Gendrel, A. V. et al. Smchd1-dependent and -independent pathways determine developmental dynamics of CpG island methylation on the inactive X chromosome. Dev. Cell 23, 265–279 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Gendrel, A.-V. et al. Epigenetic functions of smchd1 repress gene clusters on the inactive X chromosome and on autosomes. Mol. Cell. Biol. 33, 3150–3165 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Leong, H. S. et al. Epigenetic regulator Smchd1 functions as a tumor suppressor. Cancer Res. 73, 1591–1599 (2013).

    Article  PubMed  CAS  Google Scholar 

  25. Mould, A. W. et al. Smchd1 regulates a subset of autosomal genes subject to monoallelic expression in addition to being critical for X inactivation. Epigenetics Chromatin 6, 19 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Nozawa, R.-S. et al. Human inactive X chromosome is compacted through a PRC2-independent SMCHD1–HBiX1 pathway. Nat. Struct. Mol. Biol. 20, 566–573 (2013).

    Article  PubMed  CAS  Google Scholar 

  27. Mason, A. G. et al. SMCHD1 regulates a limited set of gene clusters on autosomal chromosomes. Skelet. Muscle 7, 12 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Darrow, E. M. et al. Deletion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture. Proc. Natl Acad. Sci. USA 113, E4504–E4512 (2016).

    Article  PubMed  CAS  Google Scholar 

  29. Deng, X. et al. Bipartite structure of the inactive mouse X chromosome. Genome Biol. 16, 152 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Giorgetti, L. et al. Structural organization of the inactive X chromosome in the mouse. Nature 535, 575–579 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Guo, Y. et al. CTCF/cohesin-mediated DNA looping is required for protocadherin α promoter choice. Proc. Natl Acad. Sci. USA 109, 21081–21086 (2012).

    Article  PubMed  Google Scholar 

  32. Brinkman, A. B. et al. Sequential ChIP-bisulfite sequencing enables direct genome-scale investigation of chromatin and DNA methylation cross-talk. Genome Res. 22, 1128–1138 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Saksouk, N. et al. Redundant mechanisms to form silent chromatin at pericentromeric regions rely on BEND3 and DNA methylation. Mol. Cell 56, 580–594 (2014).

    Article  PubMed  CAS  Google Scholar 

  34. Walter, M., Teissandier, A., Pérez-Palacios, R. & Bourc’his, D. An epigenetic switch ensures transposon repression upon dynamic loss of DNA methylation in embryonic stem cells. eLife 5, e11418 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  35. de Greef, J. C. et al. Smchd1 haploinsufficiency exacerbates the phenotype of a transgenic FSHD1 mouse model. Hum. Mol. Genet. 27, 716–731 (2018).

    Article  PubMed  Google Scholar 

  36. Lun, A. T. L. & Smyth, G. K. diffHic: a Bioconductor package to detect differential genomic interactions in Hi-C data. BMC Bioinformatics 16, 258 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Serra, F. et al. Automatic analysis and 3D-modelling 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 

  38. Lajoie, B. R., Dekker, J. & Kaplan, N. The Hitchhiker’s guide to Hi-C analysis: practical guidelines. Methods 72, 65–75 (2015).

    Article  PubMed  CAS  Google Scholar 

  39. 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  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Gouti, M. et al. In vitro generation of neuromesodermal progenitors reveals distinct roles for wnt signalling in the specification of spinal cord and paraxial mesoderm identity. PLoS Biol. 12, e1001937 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Gouti, M. et al. A gene regulatory network balances neural and mesoderm specification during vertebrate trunk development. Dev. Cell 41, 243–261.e7 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Lippmann, E. S. et al. Deterministic HOX patterning in human pluripotent stem cell-derived neuroectoderm. Stem Cell Rep. 4, 632–644 (2015).

    Article  CAS  Google Scholar 

  44. Wong, S. F. et al. Independent regulation of vertebral number and vertebral identity by microRNA-196 paralogs. Proc. Natl Acad. Sci. USA 112, E4884–E4893 (2015).

    Article  PubMed  CAS  Google Scholar 

  45. Wellik, D. M. Hox genes and vertebrate axial pattern. Curr. Top. Dev. Biol. 88, 257–278 (2009).

    Article  PubMed  CAS  Google Scholar 

  46. Boulet, A. M. & Capecchi, M. R. Duplication of the Hoxd11 gene causes alterations in the axial and appendicular skeleton of the mouse. Dev. Biol. 249, 96–107 (2002).

    Article  PubMed  CAS  Google Scholar 

  47. Carapuço, M., Nóvoa, A., Bobola, N. & Mallo, M. Hox genes specify vertebral types in the presomitic mesoderm. Genes Dev. 19, 2116–2121 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Akasaka, T. et al. A role for mel-18, a Polycomb group-related vertebrate gene, during theanteroposterior specification of the axial skeleton. Development 122, 1513–1522 (1996).

    PubMed  CAS  Google Scholar 

  49. Akasaka, T. et al. Mice doubly deficient for the Polycomb Group genes Mel18 and Bmi1 reveal synergy and requirement for maintenance but not initiation of Hox gene expression. Development 128, 1587–1597 (2001).

    PubMed  CAS  Google Scholar 

  50. Suzuki, M. et al. Involvement of the Polycomb-group gene Ring1B in the specification of the anterior-posterior axis in mice. Development 129, 4171–4183 (2002).

    PubMed  CAS  Google Scholar 

  51. van der Lugt, N. M. et al. Posterior transformation, neurological abnormalities, and severe hematopoietic defects in mice with a targeted deletion of the bmi-1 proto-oncogene. Genes Dev. 8, 757–769 (1994).

    Article  PubMed  Google Scholar 

  52. Royce-Tolland, M. E. et al. The A-repeat links ASF/SF2-dependent Xist RNA processing with random choice during X inactivation. Nat. Struct. Mol. Biol. 17, 948–954 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Trussart, M. et al. Defined chromosome structure in the genome-reduced bacterium Mycoplasma pneumoniae. Nat. Commun. 8, 14665 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Keniry, A. et al. Setdb1-mediated H3K9 methylation is enriched on the inactive X and plays a role in its epigenetic silencing. Epigenetics Chromatin 9, 16 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Naughton, C., Sproul, D., Hamilton, C. & Gilbert, N. Analysis of active and inactive X chromosome architecture reveals the independent organization of 30 nm and large-scale chromatin structures. Mol. Cell 40, 397–409 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Gordon, C. T. et al. De novo mutations in SMCHD1 cause Bosma arhinia microphthalmia syndrome and abrogate nasal development. Nat. Genet. 49, 249–255 (2017).

    Article  PubMed  CAS  Google Scholar 

  57. Shaw, N. D. et al. SMCHD1 mutations associated with a rare muscular dystrophy can also cause isolated arhinia and Bosma arhinia microphthalmia syndrome. Nat. Genet. 49, 238–248 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Jansz, N., Chen, K., Murphy, J. M. & Blewitt, M. E. The epigenetic regulator SMCHD1 in development and disease. Trends Genet. 33, 233–243 (2017).

    Article  PubMed  CAS  Google Scholar 

  59. Lemmers, R. J. L. F. et al. Facioscapulohumeral muscular dystrophy is uniquely associated with one of the two variants of the 4q subtelomere. Nat. Genet. 32, 235–236 (2002).

    Article  PubMed  CAS  Google Scholar 

  60. Gurzau, A. D. et al. FSHD2- and BAMS-associated mutations confer opposing effects on SMCHD1 function. J. Biol. Chem. 293, 9841–9853 (2018).

    Article  PubMed  CAS  Google Scholar 

  61. 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  PubMed  CAS  Google Scholar 

  62. Gdula, M.R. et al. The non-canonical SMC protein SmcHD1 antagonises TAD formation on the inactive X chromosome. Preprint at https://www.biorxiv.org/content/early/2018/06/08/342147 (2018).

  63. Wang, C. Y., Jégu, T., Chu, H. P., Oh, H. J. & Lee, J. T. SMCHD1 merges chromosome compartments and assists formation of super-structures on the inactive X. Cell 174, 406–421.e25 (2018).

    Article  PubMed  CAS  Google Scholar 

  64. Majewski, I. J. et al. Polycomb repressive complex 2 (PRC2) restricts hematopoietic stem cell activity. PLoS Biol. 6, e93 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Chaumeil, J., Augui, S., Chow, J. C. & Heard, E. Combined immunofluorescence, RNA fluorescent in situ hybridization, and DNA fluorescent in situ hybridization to study chromatin changes, transcriptional activity, nuclear organization, and X-chromosome inactivation. Methods Mol. Biol. 463, 297–308 (2008).

    Article  PubMed  CAS  Google Scholar 

  66. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  PubMed  CAS  Google Scholar 

  67. Gilles, J. F., Dos Santos, M., Boudier, T., Bolte, S. & Heck, N. DiAna, an ImageJ tool for object-based 3D co-localization and distance analysis. Methods 115, 55–64 (2017).

    Article  PubMed  CAS  Google Scholar 

  68. Wingett, S. et al. HiCUP: pipeline for mapping and processing Hi-C data. F1000Res. 4, 1310 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Imakaev, M. et al. Iterative correction of Hi-C data reveals hallmarks of chromosome organization. Nat. Methods 9, 999–1003 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Marco-Sola, S., Sammeth, M., Guigó, R. & Ribeca, P. The GEM mapper: fast, accurate and versatile alignment by filtration. Nat. Methods 9, 1185–1188 (2012).

    Article  PubMed  CAS  Google Scholar 

  72. 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 

  73. Needleman, S. B. & Wunsch, C. D. A general method applicable to the search for similarities in the amino acid sequence of two proteins. J. Mol. Biol. 48, 443–453 (1970).

    Article  PubMed  CAS  Google Scholar 

  74. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. 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  PubMed  CAS  Google Scholar 

  76. Wu, H. et al. Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science 329, 444–448 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Nelson, J. D., Denisenko, O. & Bomsztyk, K. Protocol for the fast chromatin immunoprecipitation (ChIP) method. Nat. Protoc. 1, 179–185 (2006).

    Article  PubMed  CAS  Google Scholar 

  78. 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 

  79. Lund, E., Oldenburg, A. R. & Collas, P. Enriched domain detector: a program for detection of wide genomic enrichment domains robust against local variations. Nucleic Acids Res. 42, e92 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Buenrostro, J., Wu, B., Chang, H. & Greenleaf, W. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  81. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Yin, D. et al. High concordance between Illumina HiSeq2500 and NextSeq500 for reduced representation bisulfite sequencing (RRBS). Genom. Data 10, 97–100 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Rigueur, D. & Lyons, K. M. Whole-mount skeletal staining. Methods Mol. Biol. 1130, 113–121 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Coughlin, R. Allan and T. Johansson for useful discussions. This work was funded by an Australian National Health and Medical Research Council grant to M.E.B., J.M.M. and M.E.R. (GNT1098290), and fellowships to J.M.M. (GNT1105754) and M.E.R. (GNT1104924). N.J. was supported by an Australian Research Training Program Fellowship. M.E.B. was supported by a Bellberry-Viertel Senior Medical Research Fellowship. The Australian Regenerative Medicine Institute is supported by grants from the State Government of Victoria and the Australian Government. This work was made possible through Victorian State Government Operational Infrastructure Support and the Australian National Health and Medical Research Council Research Institute Infrastructure Support Scheme.

Author information

Authors and Affiliations

Authors

Contributions

N.J. designed experiments, performed experiments, interpreted and analyzed data, and wrote the paper. A.K. designed experiments, performed experiments, and interpreted and analyzed data. M.T., P.H. and M.E.R. contributed to bioinformatic analyses of the data. T.B., K.B. and M.I. performed experiments. H.B. designed and performed experiments. E.M. designed experiments, performed experiments, and interpreted and analyzed data. G.F.K., I.D.T. and A.W.M. designed and performed experiments. J.M.M. designed experiments, interpreted data and edited the paper. M.E.B. designed experiments, interpreted data, and wrote and edited the paper.

Corresponding author

Correspondence to Marnie E. Blewitt.

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 In situ Hi-C-library quality-control measures.

In situ Hi-C was performed in n=3 Smchd1fl/fl and Smchd1del/del female NSC lines, one week post Cre-mediated deletion of Smchd1. a. Tapestation trace showing read-length distribution of pooled in situ HiC libraries run on a high-sensitivity D5000 Tape. Libraries were produced in the correct size range. b. These libraries were of good quality, as determined by HiCup. Pie graph from HiCup processing report representing the relative distribution of in situ HiC reads, following optimisation of restriction digest and conditions to prevent aggregation of nuclei. Valid pairs, or chimeric HiC reads, are represented in navy blue. All other colours represent a common experimental artefact. Notably the valid reads now make up 86% of the total reads. c. Multidimensional scaling (MDS) plot showing the relationship between the 3 Smchd1fl/fl and 3 Smchd1del/del in situ Hi-C samples. Samples separate by genotype on the first MDS component: Leading logFC dim1 (X axis); Leading logFC dim1: second MDS component (Y axis). d. High correlation between biological replicates indicated by a Spearman rank correlation between bins in two matrices at increasing genomic distances. e. The log interaction count (Y axis) compared to log genomic distance (X axis) shown for the 100 kb, 500 kb and 1 Mb resolution analyses. The trend line is shown for 3 different log genomic distances. These plots show the distance-dependent decay of HiC interaction frequency.

Supplementary Figure 2 In situ HiC diffHiC differential interactions and compartment analysis.

Histograms of the differential interactions (DIs) called at 100kb (a) and 1Mb (b), plotting the distance between the two genomic anchors (Bin centre, X axis) against the frequency of interactions (Y axis). c. Summary TADbit analysis using the first eigenvector to identify compartments from a correlation matrix of normalised HiC data at 1MB resolution across chromosome 11 in Smchd1fl/fl and Smchd1del/del samples, showing no significant changes in compartment assignment (90% overlap genome-wide). The first principle component of the PCA on the Hi-C correlation matrix is shown as a green line, the density as an orange line, and the blue or red block indicates A vs B compartment d-e. Distribution of strengthened (green bars) and weakened (red bars) interactions at 1 Mb (d) and 100 kb (e) resolution. Differential interactions are spread across all chromosomes, but cluster at Smchd1 binding sites such as the X chromosome.

Supplementary Figure 3 Generation of Smchd1GFP knock-in allele and its use in ChIP–seq.

a. acGFP-loxP-neomycin resistance-loxP was knocked in immediately before the stop codon of Smchd1 in 129T2/SvEms strain mESC, such that Smchd1 would be produced as a Smchd1-GFP fusion protein. Correctly targeted mESC clones were used to generate a line of mice, and the neomycin resistance cassette removed by Cre-mediated recombination. b. The Smchd1-GFP fusion protein was detected on the inactive X chromosome, as marked by H2aK119ub. GFP and H2AK119ub immunofluorescence performed in female Smchd1GFP/GFP mouse embryonic fibroblasts. c. Optimisation of the ChIP-seq sonication conditions to release the Xi from the insoluble fraction. Sonication was performed in X129Xist∆AXCastaneus NSCs. Screenshot from genome browser showing the X chromosome. Positions of genes are shown as black boxes along the top. Tracks displayed are showing normalised counts for reads that mapped to the inactive X chromosome (castaneus) minus the reads that mapped to the active X chromosome (domesticus). Height and colour of bar represents relative coverage. Sonication time is labelled on the left. As sonication time increases, reads that map to the Xi increase. Note that even after 30 minutes sonication, coverage from the Xi is still not as great as that from the Xa (relative coverage across the X chromosome is negative). Scale bar on right is read count. Data from n=1. d. Enrichment of GFP over WCE on Chromosome 2 and Chromosome 15. Height and colour of peaks indicates strength of read counts normalised to WCE; red is most enriched, blue is most depleted. HoxD locus on Chromosome 2 and HoxC locus on Chromosome 15 are marked with an asterisk. Position of MACS2-like peaks called using Seqmonk are indicated in blue.

Supplementary Figure 4 Normalized HiC interaction frequencies for interactions between the HoxB cluster and other clustered genes.

a. Normalised Hi-C interaction frequencies at 50kb, 500 kb and 1Mb resolution displayed as a heat map and rotated 45o for Chr11:96–100Mb containing the HoxB and Keratin gene clusters. Above the horizontal is the Smchd1fl/fl heatmap, below the horizontal Smchd1del/del. The colour of the contact map, from blue to red, indicates the log2(contact frequency). Dotted white/black regions indicate the region of differential interaction, which is visible in the analyses at all 3 resolutions. b. Heatmaps displayed as in a, at 100kb resolution, for the region between the Olfactory receptor cluster genes (73 Mb) and HoxB cluster (97 Mb) on chromosome 11. Between the heatmaps, genes are shown as bars (green for Olfactory receptor genes), Smchd1 ChIP-seq peaks are shown as black bars. The white boxes bound the DIs with one anchor in the HoxB cluster. The magenta boxes are all other DIs in the region.

Supplementary Figure 5 Hox genes are upregulated in Smchd1-null NMPs differentiated from mESCs.

a. Principal component analysis (PCA) of RNAseq libraries from the differentiation of Smchd1fl/fl and Smchd1del/del mESCs to NMPs. Samples separate by timepoint on the first component (45%, x-axis) and by coverage on the second (22%, y-axis). Smchd1fl/fl samples are depicted in shades of red, and Smchd1del/del samples are depicted in shades of blue. Darker colours represent early time points, whereas lighter colours represent later time points. b. Dot plots showing expression of all genes in Smchd1fl/fl (x-axis) compared with Smchd1del/del (y-axis) in log(RPM) at D1, D5, and D8 post-induction of NMP differentiation. All genes are depicted in grey. Differentially expressed genes (FDR < 0.05) are depicted in blue. Hox genes are depicted in red. Hox genes which are differentially expressed are depicted in green. Pearson’s correlation value (R) is shown on the bottom, right of each scatter plot. c. Line graphs showing Hox gene expression throughout in vitro differentiation of male Smchd1fl/fl and Smchd1del/del mESCs into NMPs, as shown in Fig. 3a. Each graph shows expression for one paralogue group. i.e. the top left graph is showing expression of the Hox1 gene in three paralogous clusters. Time in days (x axis) is plotted against log2 transformed reads per million (RPM) (y axis). Smchd1fl/fl values are plotted in blue, and Smchd1del/del in red. Circles represent genes from the HoxA cluster, squares represent HoxB, triangles represent HoxC, and upside-down triangle for those in HoxD. Solid lines depict genes that are differentially expressed (FDR < 0.05) at D8 between Smchd1fl/fl and Smchd1del/del NMP. Dotted lines indicate genes that were not called as differentially expressed using edgeR. d. Line graphs showing the downregulation of 4/5 pluripotency genes between day 1 and day 8 of the differentiation series in Smchd1fl/fl and Smchd1del/del samples.

Supplementary Figure 6 Smchd1-null embryos have altered posterior Hox-gene expression and skeletal abnormalities.

a. Images of a representative somite-matched, sex-matched pair of E9.5 embryos, Smchd1+/+ and Smchd1MommeD1/MommeD1. b. Principal component analysis (PCA) of RNA-seq libraries from the somite-matched female paired Smchd1+/+ (n=3) and Smchd1MommeD1/MommeD1 (n=4) samples. Smchd1+/+ samples are depicted in green, and Smchd1MommeD1/MommeD1 samples are depicted in blue. c. Gene expression data from somite-matched female paired Smchd1+/+ (n=3) (y-axis) against Smchd1MommeD1/MommeD1 (n=4) samples (x-axis). Genes that are differentially expressed between Smchd1+/+ and Smchd1MommeD1/MommeD1 samples (FDR<0.05) are shown in blue. The Hox genes are shown in green, X-linked DEG in red, clustered protocadherins in orange, and Smchd1 in purple. All other genes are represented in grey. Axes are mean log transformed reads per million. d-e. Representative images from n = 10 whole mount skeletons of Smchd1+/+ (d) and Smchd1MommeD1/MommeD1 (e) E17.5 embryos, stained with Alizaren red and Alician blue. Whole body image, higher magnification view of the cervical vertebrae, and higher magnification sagittal view of embryo shown in d. Smchd1MommeD1/MommeD1 embryo shows abnormal bifurcating morphology of C2, and absence of T13 rib, indicated by asterisks. Note image for C2 is not the same Smchd1MommeD1/MommeD1 embryo as full body and sagittal view. f. Summarised data from scoring of whole mount skeletons from Smchd1+/+ (n=10) and Smchd1MommeD1/MommeD1 (n=10) male embryos. Cervical, thoracic, lumbar and sacral vertebrae are indicated, with ribs shown by horizonal blue lines, sternum shown by vertical blue lines and sacrum shown by vertical red lines. Dark blue dots indicate dramatically reduced rib process. C2bif refers to a malformed C2 vertebra, where it bifurcates at the dorsal side.

Supplementary Figure 7 Smchd1 deletion does not alter the compaction accessibility of the inactive X chromosome but does result in enriched H3K27me3 on the Xi.

a. Normalised Hi-C interaction frequencies at 100 kb resolution, generated from 200 million reads randomly subsampled from libraries at each condition, displayed as a heat map for the X chromosome in Smchd1fl/fl (a) and Smchd1del/del (b) NSCs. The colour of the contact map, from blue to red, indicates the log2(contact frequency). c. ATAC-seq was performed in Smchd1fl/fl and Smchd1del/del NSCs (n=3 female, n=2 male), one week post Cre mediated deletion of Smchd1. Position of female specific X-linked differentially accessible peaks (DAP) from the ATAC-seq analysis called using and edgeR analysis on MACS2 peaks in red. Positions of genes are indicated in black along the top. d. Sex-specific differential analysis of the MACS2 peaks identified in all samples, found 540 female-specific DAPS genome-wide. These were regions that became more accessible in the absence of Smchd1 in females, but not in males, as shown by box plots showing the coverage at 36 differential X-linked ATAC-seq peaks in log (RPM) between female Smchd1fl/fl (F) and Smchd1del/del (D) NSCs (left, n=3), and males (right, n=2). The female specific DAPs were not enriched on the X chromosome relative to autosomes, X-linked DAPs comprising 6% of the total female specific DAP (36/540). p values shown were determined by Student’s two-tailed t test. e. Confocal imaging of Smchd1fl/fl (top) and Smchd1del/del (bottom) NSC nuclei following DNA FISH with X chromosome paint, n=23 observations per genotype, n=2 cell lines. DAPI stained DNA is shown (blue) and X chromosome paint (magenta), shown with 3D rendering performed in Imaris to measure volume. f. Widefield images of H3K27me3 immunofluorescence performed in female Smchd1fl/fl (top) and Smchd1del/del (bottom) NSCs (green). g. Confocal images with Airyscan processing of H3K27me3 immunofluorescence (red) in female Smchd1+/+ (top) and Smchd1MommeD1/MommeD1 NSCs (bottom). Dot plots of volume and mean intensity of focal H3K27me3 enrichment in 3D using Imaris. Statistical comparisons were made using a two-tailed student’s t-test, line indicates mean, error bars are SEM. Measurements from 17 observations in n=2 cell lines, p <0.01. h. Genome browser tracks for the H3K27me3 ChIP-seq from ChrX:63,350,830-67,476,550 on the Castaneus-derived Xi chromosome in Smchd1del/fl and Smchd1del/del NSCs (n=2). Genes are shown in black, MACS2-called peaks in Smchd1del/fl NSCs are shown in blue, and MACS2-called peaks in Smchd1del/del NSCs are shown in red. Scale is normalised ChIP-seq read counts, normalised for read depth using the total autosomal read count.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7, Supplementary Note and Supplementary Table 1

Reporting Summary

Supplementary Dataset 1

Uncropped Western blot images for Figure 1a

Supplementary Dataset 2

Differential interactions between Smchd1fl/fl and Smchd1del/del female NSCs, Smchd1fl/fl and Smchd1del/del male NSCs, based on diffHic analysis at 1 Mb, 500 kb and 100 kb resolutions

Supplementary Dataset 3

Strengthened and weakened TAD boundaries, TAD insulation scores, and compartment boundaries based on Tadbit analysis in Smchd1fl/fl and Smchd1del/del female NSCs

Supplementary Dataset 4

Smchd1-GFP ChIP-seq peaks

Supplementary Dataset 5

Loops defined by HiCCUPS in Smchd1fl/fl and Smchd1del/del female NSCs

Supplementary Dataset 6

Differentially expressed genes between Smchd1fl/fl and Smchd1del/del female NSCs

Supplementary Dataset 7

Differential ATAC-seq peaks between Smchd1fl/fl and Smchd1del/del NSCs

Supplementary Dataset 8

H3K27ac and H3K27me3 ChIP-seq peaks from Smchd1fl/fl and Smchd1del/del female NSCs

Supplementary Dataset 9

Differentially expressed genes between Smchd1fl/fl and Smchd1del/del male ES cells differentiating to neuromesodermal progenitors at days 1, 5 and 8 of differentiation

Supplementary Dataset 10

Differentially expressed genes between presomitic mesoderm from Smchd1+/+ and Smchd1MommeD1/MommeD1 E9.5 female embryos

Supplementary Dataset 11

Read counts for X-linked genes that mapped to the active X chromosome (genome 1) and the inactive X chromosome (genome 2) in Smchd1del/fl and Smchd1del/del female NSCs from RNAseq

Supplementary Dataset 12

H3K27me3 ChIP-seq peaks on the Xi from Smchd1del/fl and Smchd1del/del female NSCs

Supplementary Dataset 13

Read counts for RRBS data from the Xa and Xi of Smchd1del/fl and Smchd1del/del female NSCs

Source Data, Figure 3f

Source Data, Figure 6b, c, e, f

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jansz, N., Keniry, A., Trussart, M. et al. Smchd1 regulates long-range chromatin interactions on the inactive X chromosome and at Hox clusters. Nat Struct Mol Biol 25, 766–777 (2018). https://doi.org/10.1038/s41594-018-0111-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0111-z

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