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. Copyright © 2005 by The Endocrine Society.

Estrogen mediates phosphorylation of histone H3 in ovarian follicle and mammary epithelial tumor cells via the mitotic kinase, Aurora B / Z. T., Ruiz Cortes; S., Kimmins; Monaco, Lucia; K. H., Burns; P., Sassone Corsi; B. D., Murphy. - In: MOLECULAR ENDOCRINOLOGY. - ISSN 0888-8809. - 19:12(2005), pp. 2991-3000. [10.1210/me.2004-0441]

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

MONACO, Lucia;
2005

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. Copyright © 2005 by The Endocrine Society.
2005
01 Pubblicazione su rivista::01a Articolo in rivista
Estrogen mediates phosphorylation of histone H3 in ovarian follicle and mammary epithelial tumor cells via the mitotic kinase, Aurora B / Z. T., Ruiz Cortes; S., Kimmins; Monaco, Lucia; K. H., Burns; P., Sassone Corsi; B. D., Murphy. - In: MOLECULAR ENDOCRINOLOGY. - ISSN 0888-8809. - 19:12(2005), pp. 2991-3000. [10.1210/me.2004-0441]
File allegati a questo prodotto
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11573/68857
 Attenzione

Attenzione! I dati visualizzati non sono stati sottoposti a validazione da parte dell'ateneo

Citazioni
  • ???jsp.display-item.citation.pmc??? 10
  • Scopus 29
  • ???jsp.display-item.citation.isi??? 26
social impact