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:

Genome-wide chromatin analysis in mature mouse and human spermatozoa

A Corrigendum to this article was published on 11 March 2014

This article has been updated

Abstract

At the end of mammalian spermatogenesis, chromatin in differentiating germ cells is extensively remodeled, with the majority of nucleosomes being removed and ultimately exchanged by highly basic proteins named protamines. Residual nucleosomes are, to various degrees, retained at regulatory sequences in human and mouse sperm. Moreover, certain histone variants and modifications remain present in regulatory sequences of subsets of genes in spermatozoa, providing opportunities for paternal inheritance of chromatin states and epigenetic control of gene expression in the subsequent generation. Here we describe in detail a method that enables the generation of soluble chromatin samples from mouse and human spermatozoa within 1 d. These samples are amendable to chromatin immunoprecipitation and high-throughput sequencing of nucleosome-associated genomic DNA, which require several additional days. We also provide computational scripts that allow straightforward analysis of large genome-wide data sets by biologists with limited computational experience. This protocol will facilitate studies of mechanisms of chromatin remodeling and epigenetic reprogramming during spermatogenesis and of paternal epigenetic inheritance. Similarly, it will help in the study of the causes of human male infertility.

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

Figure 1: Schematic overview of the protocol.
Figure 2: Optimization of MNase treatment for sperm chromatin isolation (Steps 29–34).
Figure 3: Comparison of nucleosome enrichments of mouse sperm obtained by qPCR versus high-throughput sequencing analyses.
Figure 4: Analysis of H3K27me3-enriched regions in mouse sperm.
Figure 5: Reproducibility of chromatin isolation and ChIP experiments in mouse sperm.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

Change history

  • 26 February 2014

     In the version of this article initially published, the amount of N-ethylmaleimide to be dissolved was incorrect; it should be 25 mg rather than 0.25 mg. It was also not specified that the N-ethylmaleimide solution should be 1 M. In addition, the catalog number from Sigma-Aldrich for sodium acetate was incorrectly given as S2889; it should be S8625. Also, it should have been stated that the sodium acetate required is ·3H2O. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Govin, J., Caron, C., Lestrat, C., Rousseaux, S. & Khochbin, S. The role of histones in chromatin remodelling during mammalian spermiogenesis. Eur. J. Biochem. 271, 3459–3469 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Balhorn, R., Gledhill, B.L. & Wyrobek, A.J. Mouse sperm chromatin proteins: quantitative isolation and partial characterization. Biochemistry 16, 4074–4080 (1977).

    Article  CAS  PubMed  Google Scholar 

  3. Gatewood, J.M., Cook, G.R., Balhorn, R., Bradbury, E.M. & Schmid, C.W. Sequence-specific packaging of DNA in human sperm chromatin. Science 236, 962–964 (1987).

    Article  CAS  PubMed  Google Scholar 

  4. Gardiner-Garden, M., Ballesteros, M., Gordon, M. & Tam, P.P. Histone- and protamine-DNA association: conservation of different patterns within the β-globin domain in human sperm. Mol. Cell. Biol. 18, 3350–3356 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wykes, S.M. & Krawetz, S.A. The structural organization of sperm chromatin. J. Biol. Chem. 278, 29471–29477 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Pittoggi, C. et al. A fraction of mouse sperm chromatin is organized in nucleosomal hypersensitive domains enriched in retroposon DNA. J. Cell Sci. 112 (Part 20): 3537–3548 (1999).

    CAS  PubMed  Google Scholar 

  7. Arpanahi, A. et al. Endonuclease-sensitive regions of human spermatozoal chromatin are highly enriched in promoter and CTCF binding sequences. Genome Res. 19, 1338–1349 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hammoud, S.S. et al. Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Brykczynska, U. et al. Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 17, 679–687 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Erkek, S. et al. Molecular determinants of nucleosome retention at CpG-rich sequences in mouse spermatozoa. Nat. Struct. Mol. Biol. 20, 868–875 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Vavouri, T. & Lehner, B. Chromatin organization in sperm may be the major functional consequence of base composition variation in the human genome. PLoS Genet. 7, e1002036 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gill, M.E., Erkek, S. & Peters, A.H. Parental epigenetic control of embryogenesis: a balance between inheritance and reprogramming? Curr. Opin. Cell Biol. 24, 387–396 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Pembrey, M.E. et al. Sex-specific, male-line transgenerational responses in humans. Eur. J. Hum. Genet. 14, 159–166 (2006).

    Article  PubMed  Google Scholar 

  14. Kaati, G., Bygren, L.O., Pembrey, M. & Sjostrom, M. Transgenerational response to nutrition, early life circumstances and longevity. Eur. J. Hum. Genet. 15, 784–790 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Nadeau, J.H. Transgenerational genetic effects on phenotypic variation and disease risk. Hum. Mol. Genet. 18, R202–R210 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ng, S.F. et al. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature 467, 963–966 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Anway, M.D., Cupp, A.S., Uzumcu, M. & Skinner, M.K. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Chong, S. et al. Modifiers of epigenetic reprogramming show paternal effects in the mouse. Nat. Genet. 39, 614–622 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Carone, B.R. et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zeybel, M. et al. Multigenerational epigenetic adaptation of the hepatic wound-healing response. Nat. Med. 18, 1369–1377 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. van der Heijden, G.W. et al. Chromosome-wide nucleosome replacement and H3.3 incorporation during mammalian meiotic sex chromosome inactivation. Nat. Genet. 39, 251–258 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Gaucher, J. et al. From meiosis to postmeiotic events: the secrets of histone disappearance. FEBS J. 277, 599–604 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Matzuk, M.M. et al. Small-molecule inhibition of BRDT for male contraception. Cell 150, 673–684 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Goldberg, A.D. et al. Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140, 678–691 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Umlauf, D., Goto, Y. & Feil, R. Site-specific analysis of histone methylation and acetylation. Methods Mol. Biol. 287, 99–120 (2004).

    CAS  PubMed  Google Scholar 

  26. 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 

  27. Mohn, F. et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol Cell 30, 755–766 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. 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  CAS  PubMed  Google Scholar 

  29. Hublitz, P., Albert, M. & Peters, A.H. Mechanisms of transcriptional repression by histone lysine methylation. Int. J. Dev. Biol. 53, 335–354 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen 5th edn. (2010).

Download references

Acknowledgements

We gratefully acknowledge J. Hofsteenge for critical advice during the development of this protocol. We thank T. Roloff (FMI functional genomics group), I. Nissen (Laboratory for Quantitative Genomics, Department of Biosystems Science and Engineering (D-BSSE), Basel) and the FMI animal facility for excellent assistance. S.E. is supported as a recipient of a Boehringer Ingelheim Fond fellowship. Research in the Stadler and Peters laboratories is supported by the Novartis Research Foundation and the Swiss Initiative in Systems Biology (Cell Plasticity - Systems Biology of Cell Differentation). In addition, the Peters laboratory receives funding from the Swiss National Science Foundation (31003A_125386 and National Research Programme NRP63—Stem Cells and Regenerative Medicine), the Japanese Swiss Science and Technology Cooperation Program, the FP7 Marie Curie Initial Training Network “Nucleosome4D” and the EMBO Young Investigator Program.

Author information

Authors and Affiliations

Authors

Contributions

M.H., S.E., M.B.S. and A.H.F.M.P. conceived of and designed the experiments. M.H., with help of S.D.-B., developed the sperm chromatin isolation protocol, whereas S.E. and M.B.S developed the computational scripts and pipeline. L.R. contributed the purification protocol for human sperm. M.B.S. also provided bioinformatics training and support. S.E., M.H., M.B.S. and A.H.F.M.P. analyzed the data. S.E., M.B.S. and A.H.F.M.P. prepared the manuscript.

Corresponding authors

Correspondence to Michael B Stadler or Antoine H F M Peters.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Table 1

GEO identifiers (XLSX 12 kb)

Supplementary Data 1

Analysis of Next-generation-sequencing data with R (PDF 965 kb)

Supplementary Data 2

R code for analysis of next generation sequencing data (TXT 5 kb)

Supplementary Data 3

R_function_definitions_used_by_the_R_code_in_Supplementary_Files_1_and_2 (TXT 15 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hisano, M., Erkek, S., Dessus-Babus, S. et al. Genome-wide chromatin analysis in mature mouse and human spermatozoa. Nat Protoc 8, 2449–2470 (2013). https://doi.org/10.1038/nprot.2013.145

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2013.145

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