Differential in vivo binding dynamics of somatic and oocyte-specific linker histones in oocytes and during ES cell nuclear transfer.
Journal: 2005/December - Molecular Biology of the Cell
ISSN: 1059-1524
Abstract:
The embryonic genome is formed by fusion of a maternal and a paternal genome. To accommodate the resulting diploid genome in the fertilized oocyte dramatic global genome reorganizations must occur. The higher order structure of chromatin in vivo is critically dependent on architectural chromatin proteins, with the family of linker histone proteins among the most critical structural determinants. Although somatic cells contain numerous linker histone variants, only one, H1FOO, is present in mouse oocytes. Upon fertilization H1FOO rapidly populates the introduced paternal genome and replaces sperm-specific histone-like proteins. The same dynamic replacement occurs upon introduction of a nucleus during somatic cell nuclear transfer. To understand the molecular basis of this dynamic histone replacement process, we compared the localization and binding dynamics of somatic H1 and oocyte-specific H1FOO and identified the molecular determinants of binding to either oocyte or somatic chromatin in living cells. We find that although both histones associate readily with chromatin in nuclei of somatic cells, only H1FOO is capable of correct chromatin association in the germinal vesicle stage oocyte nuclei. This specificity is generated by the N-terminal and globular domains of H1FOO. Measurement of in vivo binding properties of the H1 variants suggest that H1FOO binds chromatin more tightly than somatic linker histones. We provide evidence that both the binding properties of linker histones as well as additional, active processes contribute to the replacement of somatic histones with H1FOO during nuclear transfer. These results provide the first mechanistic insights into the crucial step of linker histone replacement as it occurs during fertilization and somatic cell nuclear transfer.
Relations:
Content
Citations
(18)
References
(29)
Grants
(30)
Chemicals
(2)
Genes
(1)
Organisms
(2)
Processes
(4)
Anatomy
(6)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Mol Biol Cell 16(8): 3887-3895

Differential In Vivo Binding Dynamics of Somatic and Oocyte-specific Linker Histones in Oocytes and During ES Cell Nuclear Transfer

National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892
The Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA 19140
Department of Obstetrics and Gynaecology, Chiba University School of Medicine, Chuo-ku, Chiba 260-8670, Japan
University of Mississippi Medical School, Jackson, MS 39216
Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140
Division of Reproductive Sciences, Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT 84132
Present address: Institut fuer Medizinische Strahlenkunde und Zellforschung, University of Wuerzburg, 97078 Wuerzburg, Germany
Present address: Brown University, Providence, RI 02903.
Address correspondence to: Tom Misteli (vog.hin.liam@tiletsim).
Address correspondence to: Tom Misteli (vog.hin.liam@tiletsim).

Abstract

The embryonic genome is formed by fusion of a maternal and a paternal genome. To accommodate the resulting diploid genome in the fertilized oocyte dramatic global genome reorganizations must occur. The higher order structure of chromatin in vivo is critically dependent on architectural chromatin proteins, with the family of linker histone proteins among the most critical structural determinants. Although somatic cells contain numerous linker histone variants, only one, H1FOO, is present in mouse oocytes. Upon fertilization H1FOO rapidly populates the introduced paternal genome and replaces sperm-specific histone-like proteins. The same dynamic replacement occurs upon introduction of a nucleus during somatic cell nuclear transfer. To understand the molecular basis of this dynamic histone replacement process, we compared the localization and binding dynamics of somatic H1 and oocyte-specific H1FOO and identified the molecular determinants of binding to either oocyte or somatic chromatin in living cells. We find that although both histones associate readily with chromatin in nuclei of somatic cells, only H1FOO is capable of correct chromatin association in the germinal vesicle stage oocyte nuclei. This specificity is generated by the N-terminal and globular domains of H1FOO. Measurement of in vivo binding properties of the H1 variants suggest that H1FOO binds chromatin more tightly than somatic linker histones. We provide evidence that both the binding properties of linker histones as well as additional, active processes contribute to the replacement of somatic histones with H1FOO during nuclear transfer. These results provide the first mechanistic insights into the crucial step of linker histone replacement as it occurs during fertilization and somatic cell nuclear transfer.

Abstract

Acknowledgments

We thank Paola Scaffidi and Lisa Garrett-Beal for their assistance with microinjection experiments and Tatiana Karpova for imaging support. Imaging was done at the National Cancer Institutes Fluorescence Imaging Facility. We are grateful to Hannes Drexler for critical review of the manuscript. This study was supported in part by a grant from the National Institutes of Health/National Institute of Child Health and Human Development (HD43092) to K.L. and from the National Science Foundation to D.T.B. T.M. is a Fellow of the Keith R. Porter Endowment for Cell Biology.

Acknowledgments

Notes

This article was published online ahead of print in MBC in Press (http://www.molbiolcell.org/cgi/doi/10.1091/mbc.E05-04-0350) on June 8, 2005.

Notes
This article was published online ahead of print in MBC in Press (http://www.molbiolcell.org/cgi/doi/10.1091/mbc.E05-04-0350) on June 8, 2005.
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.