Estrogen mediates phosphorylation of histone H3 in ovarian follicle and mammary epithelial tumor cells via the mitotic kinase, Aurora B

Z. Tatiana Ruiz-Cortés, Sarah Kimmins, Lucia Monaco, Kathleen H. Burns, Paolo Sassone-Corsi, Bruce D. Murphy

Research output: Contribution to journalArticlepeer-review

25 Scopus citations

Abstract

Cells of the ovarian follicle undergo extensive proliferation and differentiation from the time that the follicle escapes from the primordial state to its acquisition of ovulatory capacity. We examined the dynamic modification of the phosphorylation state of the histone H3 N-terminal tail in granulosa cells during follicular development. In rodent follicles, the granulosa cell H3 phosphorylation on Ser10 peaks during proestrus. This epigenetic mark is induced by both FSH and 17β-estradiol (E2), acting independently. E2-induced H3 phosphorylation fails to occur in mice with inactivated α-isoform of the nuclear estrogen receptor. E2 induction of histone phosphorylation is attenuated by cell cycle inhibition. Further, E2 induces the activity of the mitotic kinase, Aurora B, in a mammary tumor cell model where mitosis is estrogen receptor-α dependent. These results provide evidence for mitotic regulation in follicle development by estrogen and demonstrate a previously undiscovered mechanism for induction of cell proliferation in ovarian and mammary gland cells.

Original languageEnglish (US)
Pages (from-to)2991-3000
Number of pages10
JournalMolecular Endocrinology
Volume19
Issue number12
DOIs
StatePublished - Dec 2005
Externally publishedYes

ASJC Scopus subject areas

  • Molecular Biology
  • Endocrinology

Fingerprint

Dive into the research topics of 'Estrogen mediates phosphorylation of histone H3 in ovarian follicle and mammary epithelial tumor cells via the mitotic kinase, Aurora B'. Together they form a unique fingerprint.

Cite this