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.

  • Protocol
  • Published:

Sister chromatid–sensitive Hi-C to map the conformation of replicated genomes

Abstract

Chromosome conformation capture (Hi-C) techniques map the 3D organization of entire genomes. How sister chromatids fold in replicated chromosomes, however, cannot be determined with conventional Hi-C because of the identical DNA sequences of sister chromatids. Here, we present a protocol for sister chromatid–sensitive Hi-C (scsHi-C) that enables the distinction of DNA contacts within individual sister chromatids (cis sister contacts) from those between sister chromatids (trans sister contacts), thereby allowing investigation of the organization of replicated genomes. scsHi-C is based on live-cell labeling of nascent DNA by the synthetic nucleoside 4-thio-thymidine (4sT), which incorporates into a distinct DNA strand on each sister chromatid because of semi-conservative DNA replication. After purification of genomic DNA and in situ Hi-C library preparation, 4sT is chemically converted into 5-methyl-cytosine in the presence of OsO4/NH4Cl to introduce T-to-C signature point mutations on 4sT-labeled DNA. The Hi-C library is then sequenced, and ligated fragments are assigned to sister chromatids on the basis of strand orientation and the presence of signature mutations. The ensemble of scsHi-C contacts thereby represents genome-wide contact probabilities within and across sister chromatids. scsHi-C can be completed in 2 weeks, has been successfully applied in HeLa cells and can potentially be established for any cell type that allows proper cell cycle synchronization and incorporation of sufficient amounts of 4sT. The genome-wide maps of replicated chromosomes detected by scsHi-C enable investigation of the molecular mechanisms shaping sister chromatid topologies and the relevance of sister chromatid conformation in crucial processes like DNA repair, mitotic chromosome formation and potentially other biological processes.

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: Genome organization throughout the cell cycle.
Fig. 2: Overview of scsHi-C methodology.
Fig. 3: Assessing 4sT toxicity in HeLa cells.
Fig. 4: Quality control measurements during scsHi-C library preparation.
Fig. 5: Genome-wide maps of sister chromatid contacts.
Fig. 6: Average contact probabilities over different genomic intervals.
Fig. 7: Average contact probability maps around sets of genomic loci.

Similar content being viewed by others

Data availability

All datasets used in the preparation of this paper were published in ref. 33 and are available from Gene Expression Omnibus under accession number GSE152373 and from the authors upon request.

Code availability

The code used to generate the figures in this manuscript was originally published in Mitter et al.33. Specifically, the ipython notebooks to generate all the plots shown in this manuscript can be found at https://github.com/gerlichlab/scshic_analysis78. A programming environment to perform all analyses shown within this manuscript is provided as a docker container at https://hub.docker.com/repository/docker/gerlichlab/scshic_docker71. The preprocessing pipeline that can be used to convert raw data to .mcool files is available at https://github.com/gerlichlab/scshic_pipeline72.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rowley, M. J. & Corces, V. G. Organizational principles of 3D genome architecture. Nat. Rev. Genet. 19, 789–800 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Finn, E. H. & Misteli, T. Molecular basis and biological function of variability in spatial genome organization. Science 365, eaaw9498 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Davidson, I. F. & Peters, J.-M. Genome folding through loop extrusion by SMC complexes. Nat. Rev. Mol. Cell Biol. 22, 445–464 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. 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  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. 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 

  10. 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 

  11. Schoenfelder, S. & Fraser, P. Long-range enhancer-promoter contacts in gene expression control. Nat. Rev. Genet. 20, 437–455 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Thiecke, M. J. et al. Cohesin-dependent and -independent mechanisms mediate chromosomal contacts between promoters and enhancers. Cell Rep. 32, 107929 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhang, Y. et al. The fundamental role of chromatin loop extrusion in physiological V(D)J recombination. Nature 573, 600–604 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ba, Z. et al. CTCF orchestrates long-range cohesin-driven V(D)J recombinational scanning. Nature 586, 305–310 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hill, L. et al. Wapl repression by Pax5 promotes V gene recombination by Igh loop extrusion. Nature 584, 142–147 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Peters, J.-M. How DNA loop extrusion mediated by cohesin enables V(D)J recombination. Curr. Opin. Cell Biol. 70, 75–83 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Naumova, N. et al. Organization of the mitotic chromosome. Science 342, 948–953 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gibcus, J. H. et al. A pathway for mitotic chromosome formation. Science 359, eaao6135 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Batty, P. & Gerlich, D. W. Mitotic chromosome mechanics: how cells segregate their genome. Trends Cell Biol. 29, 717–726 (2019).

    Article  PubMed  Google Scholar 

  20. Lupiáñez, 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 

  21. Franke, M. et al. Formation of new chromatin domains determines pathogenicity of genomic duplications. Nature 538, 265–269 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Gröschel, S. et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157, 369–381 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428–434 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. McCord, R. P., Kaplan, N. & Giorgetti, L. Chromosome conformation capture and beyond: toward an integrative view of chromosome structure and function. Mol. Cell 77, 688–708 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. 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 

  27. Belaghzal, H., Dekker, J. & Gibcus, J. H. Hi-C 2.0: an optimized Hi-C procedure for high-resolution genome-wide mapping of chromosome conformation. Methods 123, 56–65 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dekker, J., Marti-Renom, M. A. & Mirny, L. A. Exploring the three-dimensional organization of genomes: interpreting chromatin interaction data. Nat. Rev. Genet. 14, 390–403 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yatskevich, S., Rhodes, J. & Nasmyth, K. Organization of chromosomal DNA by SMC complexes. Annu. Rev. Genet. 53, 445–482 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Hustedt, N. & Durocher, D. The control of DNA repair by the cell cycle. Nat. Cell Biol. 19, 1–9 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Scully, R., Panday, A., Elango, R. & Willis, N. A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell Biol. 20, 698–714 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Goloborodko, A., Imakaev, M. V., Marko, J. F. & Mirny, L. Compaction and segregation of sister chromatids via active loop extrusion. Elife 5, e14864 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Mitter, M. et al. Conformation of sister chromatids in the replicated human genome. Nature 586, 139–144 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Herzog, V. A. et al. Thiol-linked alkylation of RNA to assess expression dynamics. Nat. Methods 14, 1198–1204 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Riml, C. et al. Osmium-mediated transformation of 4-thiouridine to cytidine as key to study RNA dynamics by sequencing. Angew. Chem. Int. Ed. Engl. 56, 13479–13483 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Lusser, A. et al. Thiouridine-to-cytidine conversion sequencing (TUC-Seq) to measure mRNA transcription and degradation rates. Methods Mol. Biol. 2062, 191–211 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Jeppsson, K., Kanno, T., Shirahige, K. & Sjögren, C. The maintenance of chromosome structure: positioning and functioning of SMC complexes. Nat. Rev. Mol. Cell Biol. 15, 601–614 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Jeppsson, K. et al. The chromosomal association of the Smc5/6 complex depends on cohesion and predicts the level of sister chromatid entanglement. PLoS Genet. 10, e1004680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kadyk, L. C. & Hartwell, L. H. Sister chromatids are preferred over homologs as substrates for recombinational repair in Saccharomyces cerevisiae. Genetics 132, 387–402 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liang, F., Han, M., Romanienko, P. J. & Jasin, M. Homology-directed repair is a major double-strand break repair pathway in mammalian cells. Proc. Natl Acad. Sci. USA. 95, 5172–5177 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Aymard, F. et al. Transcriptionally active chromatin recruits homologous recombination at DNA double-strand breaks. Nat. Struct. Mol. Biol. 21, 366–374 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Clouaire, T. et al. Comprehensive mapping of histone modifications at DNA double-strand breaks deciphers repair pathway chromatin signatures. Mol. Cell 72, 250–262.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Schep, R. et al. Impact of chromatin context on Cas9-induced DNA double-strand break repair pathway balance. Mol. Cell 81, 2216–2230.e10 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sjögren, C. & Nasmyth, K. Sister chromatid cohesion is required for postreplicative double-strand break repair in Saccharomyces cerevisiae. Curr. Biol. 11, 991–995 (2001).

    Article  PubMed  Google Scholar 

  45. Potts, P. R., Porteus, M. H. & Yu, H. Human SMC5/6 complex promotes sister chromatid homologous recombination by recruiting the SMC1/3 cohesin complex to double-strand breaks. EMBO J. 25, 3377–3388 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Watrin, E. & Peters, J.-M. Cohesin and DNA damage repair. Exp. Cell Res. 312, 2687–2693 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Waizenegger, I. C., Hauf, S., Meinke, A. & Peters, J. M. Two distinct pathways remove mammalian cohesin from chromosome arms in prophase and from centromeres in anaphase. Cell 103, 399–410 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Losada, A., Hirano, M. & Hirano, T. Cohesin release is required for sister chromatid resolution, but not for condensin-mediated compaction, at the onset of mitosis. Genes Dev. 16, 3004–3016 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hirota, T., Gerlich, D., Koch, B., Ellenberg, J. & Peters, J.-M. Distinct functions of condensin I and II in mitotic chromosome assembly. J. Cell Sci. 117, 6435–6445 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Gerlich, D., Hirota, T., Koch, B., Peters, J.-M. & Ellenberg, J. Condensin I stabilizes chromosomes mechanically through a dynamic interaction in live cells. Curr. Biol. 16, 333–344 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Zhiteneva, A. et al. Mitotic post-translational modifications of histones promote chromatin compaction in vitro. Open Biol. 7, 170076 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ginno, P. A., Burger, L., Seebacher, J., Iesmantavicius, V. & Schübeler, D. Cell cycle-resolved chromatin proteomics reveals the extent of mitotic preservation of the genomic regulatory landscape. Nat. Commun. 9, 4048 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhang, H. et al. Chromatin structure dynamics during the mitosis-to-G1 phase transition. Nature 576, 158–162 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Abramo, K. et al. A chromosome folding intermediate at the condensin-to-cohesin transition during telophase. Nat. Cell Biol. 21, 1393–1402 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kempfer, R. & Pombo, A. Methods for mapping 3D chromosome architecture. Nat. Rev. Genet. 21, 207–226 (2020).

    Article  CAS  PubMed  Google Scholar 

  56. Jerkovic, I. & Cavalli, G. Understanding 3D genome organization by multidisciplinary methods. Nat. Rev. Mol. Cell Biol. 22, 511–528 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Nagasaka, K., Hossain, M. J., Roberti, M. J., Ellenberg, J. & Hirota, T. Sister chromatid resolution is an intrinsic part of chromosome organization in prophase. Nat. Cell Biol. 18, 692–699 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. Falconer, E. et al. Identification of sister chromatids by DNA template strand sequences. Nature 463, 93–97 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Stanyte, R. et al. Dynamics of sister chromatid resolution during cell cycle progression. J. Cell Biol. 217, 1985–2004 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Oomen, M. E., Hedger, A. K., Watts, J. K. & Dekker, J. Detecting chromatin interactions between and along sister chromatids with SisterC. Nat. Methods 17, 1002–1009 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mitter, M. & Gerlich, D. W. Mapping sister chromatid conformation in replicated chromosomes. Trends Biochem. Sci. 46, 169–170 (2021).

    Article  CAS  PubMed  Google Scholar 

  63. Mumbach, M. R. et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    Article  CAS  PubMed  Google Scholar 

  65. Holland, A. J., Fachinetti, D., Han, J. S. & Cleveland, D. W. Inducible, reversible system for the rapid and complete degradation of proteins in mammalian cells. Proc. Natl Acad. Sci. USA. 109, E3350–E3357 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. 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 

  67. Yesbolatova, A. et al. The auxin-inducible degron 2 technology provides sharp degradation control in yeast, mammalian cells, and mice. Nat. Commun. 11, 5701 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kurtzer, G. M., Sochat, V. & Bauer, M. W. Singularity: scientific containers for mobility of compute. PLoS One 12, e0177459 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Di Tommaso, P. et al. Nextflow enables reproducible computational workflows. Nat. Biotechnol. 35, 316–319 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Abdennur, N. & Mirny, L. A. Cooler: scalable storage for Hi-C data and other genomically labeled arrays. Bioinformatics 36, 311–316 (2020).

    Article  CAS  PubMed  Google Scholar 

  71. Langer, C. C. H. & Mitter, M. Container with Tools to Analyze scsHi-C Data. Available at https://zenodo.org/record/5743325#.YgsVdO7MK8U (2021).

  72. Langer, C. C. H. scsHi-C Preprocessing Nextflow Pipeline (2021); https://zenodo.org/record/5742764#.YhYHEejMJZc

  73. Mitter, M. & Langer, C. C. H. A Collection of NGS Analysis Tools. Available at https://zenodo.org/record/5742702#.YgsWI-7MK8U (2021).

  74. Mitter, M. & Langer, C. C. H. HiglassUp: A higlass Upload Tool. Available at https://zenodo.org/record/5743331#.YgsWee7MK8U (2021).

  75. Held, M. et al. CellCognition: time-resolved phenotype annotation in high-throughput live cell imaging. Nat. Methods 7, 747–754 (2010).

    Article  CAS  PubMed  Google Scholar 

  76. Sommer, C., Hoefler, R., Samwer, M. & Gerlich, D. W. A deep learning and novelty detection framework for rapid phenotyping in high-content screening. Mol. Biol. Cell 28, 3428–3436 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Hande, K. R. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 34, 1514–1521 (1998).

    Article  CAS  PubMed  Google Scholar 

  78. Mitter, M. & Langer, C. C. H. scsHi-C Analysis Notebooks. Available at https://zenodo.org/record/5742704#.YgsW2u7MK8U (2021).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. The HDF Group. Hierarchical Data Format Version 5. Available at http://www.hdfgroup.org/HDF5 (2006).

  81. Kerpedjiev, P. et al. HiGlass: web-based visual exploration and analysis of genome interaction maps. Genome Biol. 19, 125 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  83. Erceg, J. et al. The genome-wide multi-layered architecture of chromosome pairing in early Drosophila embryos. Nat. Commun. 10, 1–13 (2019).

    Article  CAS  Google Scholar 

  84. Juicer and Juicebox for chromatin conformation analysis. Nat. Methods 13, 816 (2016).

  85. An, L. et al. OnTAD: hierarchical domain structure reveals the divergence of activity among TADs and boundaries. Genome Biol. 20, 282 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Yang, T. et al. HiCRep: assessing the reproducibility of Hi-C data using a stratum-adjusted correlation coefficient. Genome Res. 27, 1939–1949 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Gassler, J. et al. A mechanism of cohesin-dependent loop extrusion organizes zygotic genome architecture. EMBO J. 36, 3600–3618 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. 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 

Download references

Acknowledgements

The authors acknowledge technical support by the IMBA/IMP/GMI BioOptics and Molecular Biology Services facilities and the Vienna BioCenter Metabolomics and Next Generation Sequencing facilities. Research in the laboratory of D.W.G. is supported by the Austrian Academy of Sciences, the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. 101019039), the Austrian Science Fund (FWF; Doktoratskolleg ‘Chromosome Dynamics’ DK W1238) and the Vienna Science and Technology Fund (WWTF; projects LS17-003 and LS19-001). Research in the laboratory of R.M. is supported by the Austrian Science Fund (P31691 and F8011), the Austrian Research Promotion Agency FFG (West-Austrian BioNMR 858017) and the WWTF (project nr. LS17-003). M.M. received a PhD fellowship from the Boehringer Ingelheim Fonds. Z.T. received a Hertha Firnberg Programme fellowship of the FWF (T 1246). The VBCF Metabolomics Facility is funded by the City of Vienna through the Vienna Business Agency.

Author information

Authors and Affiliations

Authors

Contributions

M.M. developed the protocol for scsHi-C, with help from R.M. (4sT conversion chemistry), T.K. (mass spectrometry), C.C.H.L. (data processing) and D.W.G. (biological interpretation). M.M., Z.T. and D.W.G. wrote the manuscript, except the procedures section on mass spectrometry, which was written by T.K. D.W.G., M.M., Z.T. and R.M. acquired funding.

Corresponding authors

Correspondence to Michael Mitter or Daniel W. Gerlich.

Ethics declarations

Competing interests

R.M. is listed as inventor on a patent application that has been filed concerning the nucleoside conversion chemistry of this work (Osmiumtetroxide-based conversion of RNA and DNA containing thiolated nucleotides, US Patent App. 16/533,988). The other authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Feng Yue and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Related links

Key reference using this protocol

Mitter, M. et al. Nature 586, 139–144 (2020): https://doi.org/10.1038/s41586-020-2744-4

Extended data

Extended Data Fig. 1 Yield of scsHi-C using HeLa Kyoto cells and 4sT incorporation in other cell lines.

a, Yield of scsHi-C performed by using HeLa Kyoto cells at different steps of the scsHi-C protocol. b, Quantification of 4sT incorporation into genomic DNA of HCT116, HEK293 and RPE1 cells. Cells were cultured in the presence of 2 mM 4sT for 5 d, and genomic DNA was purified, digested into nucleosides and analyzed by mass spectrometry. The percentage of 4sT over total thymidine is shown from n = 2 biologically independent experiments for each cell line.

Extended Data Fig. 2 HPLC-tandem MS chromatograms of a separated standard mixture of 4sT and thymidine.

Top panels, SRM (selected reaction monitoring) traces of dT (m/z 243.1 to m/z 127.1) and of its in-source fragmentation product (m/z 127 to m/z 54) are shown. Lower panels, SRM traces of thio-dT (m/z 259.1 to m/z 143.1) and the respective in-source fragmentation product (m/z 127.1 to m/z 54.1) are depicted. Both nucleosides generate only one fragment ion (neutral loss of the sugar); therefore, we recommend recording the SRM traces of the in-source products, which can either be used for quantification (if of significant signal intensity) or as a qualifier for confirming the respective nucleoside.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mitter, M., Takacs, Z., Köcher, T. et al. Sister chromatid–sensitive Hi-C to map the conformation of replicated genomes. Nat Protoc 17, 1486–1517 (2022). https://doi.org/10.1038/s41596-022-00687-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-022-00687-6

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing