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:

Hi-CO: 3D genome structure analysis with nucleosome resolution

Abstract

The nucleosome is the basic organizational unit of the genome. The folding structure of nucleosomes is closely related to genome functions, and has been reported to be in dynamic interplay with binding of various nuclear proteins to genomic loci. Here, we describe our high-throughput chromosome conformation capture with nucleosome orientation (Hi-CO) technology to derive 3D nucleosome positions with their orientations at every genomic locus in the nucleus. This technology consists of an experimental procedure for nucleosome proximity analysis and a computational procedure for 3D modeling. The experimental procedure is based on an improved method of high-throughput chromosome conformation capture (Hi-C) analysis. Whereas conventional Hi-C allows spatial proximity analysis among genomic loci with 1–10 kbp resolution, our Hi-CO allows proximity analysis among DNA entry or exit points at every nucleosome locus. This analysis is realized by carrying out ligations among the entry/exit points in every nucleosome in a micrococcal-nuclease-fragmented genome, and by quantifying frequencies of ligation products with next-generation sequencing. Our protocol has enabled this analysis by cleanly excluding unwanted non-ligation products that are abundant owing to the frequent genome fragmentation by micrococcal nuclease. The computational procedure is based on simulated annealing-molecular dynamics, which allows determination of optimized 3D positions and orientations of every nucleosome that satisfies the proximity ligation data sufficiently well. Typically, examination of the Saccharomyces cerevisiae genome with 130 million sequencing reads facilitates analysis of a total of 66,360 nucleosome loci with 6.8 nm resolution. The technique requires 2–3 weeks for sequencing library preparation and 2 weeks for simulation.

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

Fig. 1: An overview of the Hi-CO procedure.
Fig. 2: A workflow of the sequenced read analysis to derive the Hi-CO matrix.
Fig. 3: Optimization of the MNase digestion step.
Fig. 4: Schematic representation of the DNA adaptors.
Fig. 5: Excision of a gel piece containing the ligation products.
Fig. 6: Size distribution of the DNA library.
Fig. 7: The nucleosome model for the SA-MD simulation.
Fig. 8: Trends in probabilities of read pairs as a function of genomic distance.
Fig. 9: Typical Hi-CO contact matrix.
Fig. 10: 3D nucleosome folding structure.

Similar content being viewed by others

Data availability

All data presented in this study are generated using the raw data shown in our previous publication12 that are available at BioProject accession number PRJNA427106. Source data are provided with this paper.

Code availability

Software for SA-MD simulation and a LabVIEW code for analyzing paired-end sequencing reads to output the Hi-CO matrix are available at https://doi.org/10.6084/m9.figshare.13176101.v1. Sample Hi-CO data measuring nucleosome contacts for chromosome 1 of the S. cerevisiae genome are available at the same link.

References

  1. Kornberg, R. D. Chromatin structure: a repeating unit of histones and DNA. Science 184, 868–871 (1974).

    CAS  PubMed  Google Scholar 

  2. Kornberg, R. D. & Lorch, Y. Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome. Cell 98, 285–294 (1999).

    Article  CAS  PubMed  Google Scholar 

  3. Ohno, M., Priest, D. G. & Taniguchi, Y. Nucleosome-level 3D organization of the genome. Biochem. Soc. Trans. 46, 491–501 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Baldi, S., Korber, P. & Becker, P. B. Beads on a string—nucleosome array arrangements and folding of the chromatin fiber. Nat. Struct. Biol. 27, 109–118 (2020).

    Article  CAS  Google Scholar 

  5. Li, G. et al. Highly compacted chromatin formed in vitro reflects the dynamics of transcription activation in vivo. Mol. Cell 38, 41–53 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Ou, H. D. et al. ChromEMT: visualizing 3D chromatin structure and compaction in interphase and mitotic cells. Science 357, eaag0025 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. 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 

  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. Ma, W. et al. Fine-scale chromatin interaction maps reveal the cis-regulatory landscape of human lincRNA genes. Nat. Methods 12, 71–78 (2015).

    Article  PubMed  CAS  Google Scholar 

  12. Ohno, M. et al. Sub-nucleosomal genome structure reveals distinct nucleosome folding motifs. Cell 176, 520–534.e25 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Jin, F. et al. A high-resolution map of the three-dimensional chromatin interactome in human cells. Nature 503, 290–294 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hsieh, T.-H. S. et al. Mapping nucleosome resolution chromosome folding in yeast by Micro-C. Cell 162, 108–119 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hsieh, T.-H. S., Fudenberg, G., Goloborodko, A., Rando, O. J. & Micro-C, X. L. assaying chromosome conformation from the nucleosome to the entire genome. Nat. Methods 13, 1009–1011 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. Quinodoz, S. A. et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 174, 744–757.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zheng, M. et al. Multiplex chromatin interactions with single-molecule precision. Nature 566, 558–562 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ulahannan, N., Pendleton, M., Deshpande, A. & Schwenk, S. Nanopore sequencing of DNA concatemers reveals higher-order features of chromatin structure. Preprint at bioRxiv https://doi.org/10.1101/833590 (2019)

  20. Hua, N. et al. Producing genome structure populations with the dynamic and automated PGS software. Nat. Protoc. 13, 915–926 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Zhou, X., Blocker, A. W., Airoldi, E. M. & O'Shea, E. K. A computational approach to map nucleosome positions and alternative chromatin states with base pair resolution. eLife 5, e16970 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Kubik, S. et al. Nucleosome stability distinguishes two different promoter types at all protein-coding genes in yeast. Mol. Cell 60, 422–434 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Wal, M. & Pugh, B. F. Genome-wide mapping of nucleosome positions in yeast using high-resolution MNase ChIP-Seq. Methods Enzymol. 513, 233–250 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Henikoff, J. G., Belsky, J. A., Krassovsky, K., MacAlpine, D. M. & Henikoff, S. Epigenome characterization at single base-pair resolution. Proc. Natl Acad. Sci. USA 108, 18318–18323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. McKnight, L. E. et al. Rapid and inexpensive preparation of genome-wide nucleosome footprints from model and non-model organisms. Preprint at bioRxiv https://doi.org/10.1101/870659 (2019).

  27. Tsankov, A. M. et al. Transcription factor binding dynamics during human ES cell differentiation. Nature 518, 344–349 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Skene, P. J. & Henikoff, S. A simple method for generating high-resolution maps of genome-wide protein binding. eLife 4, e09225 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Bao, Y. et al. Nucleosomes containing the histone variant H2A.Bbd organize only 118 base pairs of DNA. EMBO J 23, 3314–3324 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cao, S., Zhou, K., Zhang, Z., Luger, K. & Straight, A. F. Constitutive centromere-associated network contacts confer differential stability on CENP-A nucleosomes in vitro and in the cell. Mol. Biol. Cell 29, 751–762 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Brehove, M. et al. Histone core phosphorylation regulates DNA accessibility. J. Biol. Chem. 290, 22612–22621 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Krajewski, W. A., Li, J. & Dou, Y. Effects of histone H2B ubiquitylation on the nucleosome structure and dynamics. Nucleic Acids Res. 46, 7631–7642 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Luo, D. et al. MNase, as a probe to study the sequence-dependent site exposures in the+ 1 nucleosomes of yeast. Nucleic Acids Res. 46, 7124–7137 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Horike, S.-I., Cai, S., Miyano, M., Cheng, J.-F. & Kohwi-Shigematsu, T. Loss of silent-chromatin looping and impaired imprinting of DLX5 in Rett syndrome. Nat. Genet. 37, 31–40 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Weiner, A. et al. High-resolution chromatin dynamics during a yeast stress response. Mol. Cell 58, 371–386 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Duan, Z. et al. A three-dimensional model of the yeast genome. Nature 465, 363–367 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fullwood, M. J. et al. An oestrogen-receptor-alpha-bound human chromatin interactome. Nature 462, 58–64 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kalhor, R., Tjong, H., Jayathilaka, N., Alber, F. & Chen, L. Genome architectures revealed by tethered chromosome conformation capture and population-based modeling. Nat. Biotechnol. 30, 90–98 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Li, H. et al. Genome Project Data Processing Subgroup. 2009. The Sequence alignment/map (SAM) format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  40. Barbi, M., Mozziconacci, J., Wong, H. & Victor, J.-M. DNA topology in chromosomes: a quantitative survey and its physiological implications. J. Math. Biol. 68, 145–179 (2014).

    Article  PubMed  Google Scholar 

  41. Luger, K., Mäder, A. W., Richmond, R. K., Sargent, D. F. & Richmond, T. J. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 389, 251–260 (1997).

    Article  CAS  PubMed  Google Scholar 

  42. Richmond, T. J. & Davey, C. A. The structure of DNA in the nucleosome core. Nature 423, 145–150 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Hagerman, P. J. Flexibility of DNA. Annu. Rev. Biophys. Biophys. Chem. 17, 265–286 (1988).

    Article  CAS  PubMed  Google Scholar 

  44. Lesne, A., Riposo, J., Roger, P., Cournac, A. & Mozziconacci, J. 3D genome reconstruction from chromosomal contacts. Nat. Methods 11, 1141–1143 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Leach, A. R. Molecular Modelling: Principles and Applications (Pearson Education, 2001).

  46. Leimkuhler, B. & Matthews, C. Robust and efficient configurational molecular sampling via Langevin dynamics. J. Chem. Phys. 138, 174102 (2013).

    Article  PubMed  CAS  Google Scholar 

  47. Amberg, D. C., Burke, D. & Strathern, J. N. Methods in Yeast Genetics: A Cold Spring Harbor Laboratory Course Manual (CSHL Press, 2005).

  48. Zhang, J. et al. ChIA-PET analysis of transcriptional chromatin interactions. Methods 58, 289–299 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Dekker, J. Mapping in vivo chromatin interactions in yeast suggests an extended chromatin fiber with regional variation in compaction. J. Biol. Chem. 283, 34532–34540 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996).

    CAS  PubMed  Google Scholar 

  51. Venkatesh, S. & Workman, J. L. Histone exchange, chromatin structure and the regulation of transcription. Nat. Rev. Mol. Cell Biol. 16, 178–189 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by PRESTO; Japan Science and Technology Agency (JPMJPR15F7); grants-in-aid for Scientific Research (A) (20H00460), Challenging Exploratory Research (26650055), Challenging Pioneering Research (19H05545 and 20K20458) and Scientific Research on Innovative Areas (23115005); Japan Society for the Promotion of Science; and grants from the RIKEN Epigenome Manipulation Project, RIKEN Incentive Research Projects, Astellas Foundation for Research on Metabolic Disorders, Suntory Rising Stars Encouragement Program in Life Sciences (SunRiSE), the Takeda Science Foundation, and the Mochida Memorial Foundation for Medical and Pharmaceutical Research. D.P. acknowledges additional support from a RIKEN Foreign Postdoctoral Researcher fellowship.

Author information

Authors and Affiliations

Authors

Contributions

M.O. developed the experimental protocol. M.O. and Y.T. developed the sequence analysis method. M.O., T.A. and Y.T. developed the simulation method. M.O., T.A., D.G.P. and Y.T. wrote the manuscript.

Corresponding author

Correspondence to Yuichi Taniguchi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Related links

Key reference using this protocol

Ohno, M. et al. Cell 176, 520–534 (2019): https://doi.org/10.1016/j.cell.2018.12.014

Extended data

Extended Data Fig. 1 DNA fragments from input and ChIP samples.

ag, Acrylamide gel images of MNase-digested genomes before (input) and after (ChIP) immunoprecipitation, in a replicate experiment for Fig. 3b using histone H3 antibody (a), or experiments using antibodies against other core histones (H2B and H4) (b), using histone H4 antibody at different MNase digestion conditions (c), using a GFP antibody for analyzing a strain containing GFP-fused histone H4 at different cell cycle phases (df) and using a his-tag antibody for analyzing a strain expressing his-tag-fused histone H4 (g). In c, the arrows indicate the conditions described in the protocol.

Extended Data Fig. 2 Hi-CO matrix with colors swapped.

a,b, In addition to a Hi-CO matrix with the original color scheme (a), a matrix where the cyan and yellow color are swapped (b) is shown. Data at YDL127W gene locus are used to generate the matrix.

Supplementary information

Source data

Source Data Fig. 8

Raw data of probability graph in a.

Source Data Fig. 9

Raw data of contact matrix.

Source Data Fig. 10

Raw data of XYZ coordinates of nucleosome models.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ohno, M., Ando, T., Priest, D.G. et al. Hi-CO: 3D genome structure analysis with nucleosome resolution. Nat Protoc 16, 3439–3469 (2021). https://doi.org/10.1038/s41596-021-00543-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00543-z

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