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.

  • Letter
  • Published:

The histone H3.3 chaperone HIRA is essential for chromatin assembly in the male pronucleus

Abstract

In sexually reproducing animals, a crucial step in zygote formation is the decondensation of the fertilizing sperm nucleus into a DNA replication-competent male pronucleus. Genome-wide nucleosome assembly on paternal DNA implies the replacement of sperm chromosomal proteins, such as protamines, by maternally provided histones1,2. This fundamental process is specifically impaired in sésame (ssm), a unique Drosophila maternal effect mutant that prevents male pronucleus formation3. Here we show that ssm is a point mutation in the Hira gene, thus demonstrating that the histone chaperone protein HIRA is required for nucleosome assembly during sperm nucleus decondensation. In vertebrates, HIRA has recently been shown to be critical for a nucleosome assembly pathway independent of DNA synthesis that specifically involves the H3.3 histone variant4,5. We also show that nucleosomes containing H3.3, and not H3, are specifically assembled in paternal Drosophila chromatin before the first round of DNA replication. The exclusive marking of paternal chromosomes with H3.3 represents a primary epigenetic distinction between parental genomes in the zygote, and underlines an important consequence of the critical and highly specialized function of HIRA at fertilization.

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: Hira is the ssm gene.
Figure 2: Maternal HIRA accumulates in the male nucleus.
Figure 3: H3.3 is specifically deposited in paternal chromatin.
Figure 4: Di- or tri-methylation of H3 on lysine 4 and lysine 9 marks maternal chromosomes.

Similar content being viewed by others

References

  1. Wright, S. J. Sperm nuclear activation during fertilization. Curr. Top. Dev. Biol. 46, 133–178 (1999)

    Article  CAS  Google Scholar 

  2. McLay, D. W. & Clarke, H. J. Remodelling the paternal chromatin at fertilization in mammals. Reproduction 125, 625–633 (2003)

    Article  CAS  Google Scholar 

  3. Loppin, B., Docquier, M., Bonneton, F. & Couble, P. The maternal effect mutation sésame affects the formation of the male pronucleus in Drosophila melanogaster. Dev. Biol. 222, 392–404 (2000)

    Article  CAS  Google Scholar 

  4. Ray-Gallet, D. et al. HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol. Cell 9, 1091–1100 (2002)

    Article  CAS  Google Scholar 

  5. Tagami, H., Ray-Gallet, D., Almouzni, G. & Nakatani, Y. Histone H3.1 and H3.3 complexes mediate nucleosome assembly pathways dependent or independent of DNA synthesis. Cell 116, 51–61 (2004)

    Article  CAS  Google Scholar 

  6. Loppin, B., Berger, F. & Couble, P. The Drosophila maternal gene sésame is required for sperm chromatin remodeling at fertilization. Chromosoma 110, 430–440 (2001)

    Article  CAS  Google Scholar 

  7. Jayaramaiah Raja, S. & Renkawitz-Pohl, R. Replacement by Drosophila melanogaster protamines and Mst77F of histones during chromatin condensation in late spermatids and role of Sésame in the removal of these proteins from the male pronucleus. Mol. Cell. Biol. 25, 6165–6177 (2005)

    Article  Google Scholar 

  8. Loppin, B. & Karr, T. L. in Comprehensive Molecular Insect Science (eds Gilbert, L. B. & Iatrou, K.) 213–236 (Elsevier, Oxford, 2004)

    Google Scholar 

  9. Llevadot, R. et al. Cloning, chromosome mapping and expression analysis of the HIRA gene from Drosophila melanogaster. Biochem. Biophys. Res. Commun. 249, 486–491 (1998)

    Article  CAS  Google Scholar 

  10. Kirov, N., Shtilbans, A. & Rushlow, C. Isolation and characterization of a new gene encoding a member of the HIRA family of proteins from Drosophila melanogaster. Gene 212, 323–332 (1998)

    Article  CAS  Google Scholar 

  11. Smith, T. F., Gaitatzes, C., Saxena, K. & Neer, E. J. The WD repeat: a common architecture for diverse functions. Trends Biochem. Sci. 24, 181–185 (1999)

    Article  CAS  Google Scholar 

  12. Foe, V. E., Odell, G. M. & Edgar, B. A. in The Development of Drosophila melanogaster (eds Bates, M. & Martinez Arias, A.) 149–300 (Cold Spring Harbor Press, Cold Spring Harbor, New York, 1993)

    Google Scholar 

  13. Ahmad, K. & Henikoff, S. Histone H3 variants specify modes of chromatin assembly. Proc. Natl Acad. Sci. USA 99 (suppl.), 16477–16484 (2002)

    Article  ADS  CAS  Google Scholar 

  14. Ahmad, K. & Henikoff, S. The histone variant H3.3 marks active chromatin by replication-independent nucleosome assembly. Mol. Cell 9, 1191–1200 (2002)

    Article  CAS  Google Scholar 

  15. Janicki, S. M. et al. From silencing to gene expression: real-time analysis in single cells. Cell 116, 683–698 (2004)

    Article  CAS  Google Scholar 

  16. Schwartz, B. E. & Ahmad, K. Transcriptional activation triggers deposition and removal of the histone variant H3.3. Genes Dev. 19, 804–814 (2005)

    Article  CAS  Google Scholar 

  17. Henikoff, S., Furuyama, T. & Ahmad, K. Histone variants, nucleosome assembly and epigenetic inheritance. Trends Genet. 20, 320–326 (2004)

    Article  CAS  Google Scholar 

  18. Akhmanova, A. S. et al. Structure and expression of histone H3.3 genes in Drosophila melanogaster and Drosophila hydei. Genome 38, 586–600 (1995)

    Article  CAS  Google Scholar 

  19. Yamaguchi, M., Date, T. & Matsukage, A. Distribution of PCNA in Drosophila embryo during nuclear division cycles. J. Cell Sci. 100, 729–733 (1991)

    PubMed  Google Scholar 

  20. Roberts, C. et al. Targeted mutagenesis of the Hira gene results in gastrulation defects and patterning abnormalities of mesoendodermal derivatives prior to early embryonic lethality. Mol. Cell. Biol. 22, 2318–2328 (2002)

    Article  CAS  Google Scholar 

  21. van der Heijden, G. W. et al. Asymmetry in Histone H3 variants and lysine methylation between paternal and maternal chromatin of the early mouse zygote. Mech. Dev. 122, 1008–1022 (2005)

    Article  CAS  Google Scholar 

  22. Cowell, I. G. et al. Heterochromatin, HP1 and methylation at lysine 9 of histone H3 in animals. Chromosoma 111, 22–36 (2002)

    Article  CAS  Google Scholar 

  23. Arney, K. L., Bao, S., Bannister, A. J., Kouzarides, T. & Surani, M. A. Histone methylation defines epigenetic asymmetry in the mouse zygote. Int. J. Dev. Biol. 46, 317–320 (2002)

    CAS  PubMed  Google Scholar 

  24. Lepikhov, K. & Walter, J. Differential dynamics of histone H3 methylation at positions K4 and K9 in the mouse zygote. BMC Dev. Biol. 4, 12 (2004)

    Article  Google Scholar 

  25. Mayer, W., Niveleau, A., Walter, J., Fundele, R. & Haaf, T. Demethylation of the zygotic paternal genome. Nature 403, 501–502 (2000)

    Article  ADS  CAS  Google Scholar 

  26. Huynh, K. D. & Lee, J. T. Inheritance of a pre-inactivated paternal X chromosome in early mouse embryos. Nature 426, 857–862 (2003)

    Article  ADS  CAS  Google Scholar 

  27. Okamoto, I., Otte, A. P., Allis, C. D., Reinberg, D. & Heard, E. Epigenetic dynamics of imprinted X inactivation during early mouse development. Science 303, 644–649 (2004)

    Article  ADS  CAS  Google Scholar 

  28. Klemenz, R., Weber, U. & Gehring, W. J. The white gene as a marker in a new P-element vector for gene transfer in Drosophila. Nucleic Acids Res. 15, 3947–3959 (1987)

    Article  CAS  Google Scholar 

  29. Drysdale, R. A., Crosby, M. A. & The FlyBase Consortium. FlyBase: genes and gene models. Nucleic Acids Res. 33 (database issue), D390–D395 (2005).

Download references

Acknowledgements

We thank P. Fisher for anti-PCNA antibodies and the European Drosophila Genome Project for cosmid clones. We are grateful to B. Durand and B. Horard for helpful discussions. We also thank J. Schmitt and the CTµ microscopy center for technical assistance. This work was supported by the C.N.R.S. and the French Ministry of Research.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Benjamin Loppin.

Ethics declarations

Competing interests

Reprints and permissions information is available at npg.nature.com/reprintsandpermissions. The authors declare no competing financial interests.

Supplementary information

Supplementary Figure 1

RT-PCR analysis of Hira expression (PDF 735 kb)

Supplementary Figure 2

Western Blot analysis of HIRA-FLAG, H3.3-FLAG and H3-FLAG proteins. (PDF 283 kb)

Supplementary Figure 3

Pronuclei replicate their DNA by the time they appose. (PDF 532 kb)

Supplementary Table 1

Complementation of the ssm185b phenotype with Hira transgenes. (PDF 783 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Loppin, B., Bonnefoy, E., Anselme, C. et al. The histone H3.3 chaperone HIRA is essential for chromatin assembly in the male pronucleus. Nature 437, 1386–1390 (2005). https://doi.org/10.1038/nature04059

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature04059

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