Alterations in Brca1 expression in mouse ovarian granulosa cells have short-term and long-term consequences on estrogen-responsive organs

Lab Invest. 2012 Jun;92(6):802-11. doi: 10.1038/labinvest.2012.58. Epub 2012 Apr 9.

Abstract

Incessant menstrual cycle activity, uninterrupted by either pregnancy or oral contraceptive use, is the most important risk factor for sporadic ovarian cancer. Menstrual cycle progression is partly controlled by steroid hormones such as estrogens and others that are secreted by the ovarian granulosa cells. We showed earlier that mice carrying a homozygous granulosa cell-specific knockout of Brca1, the homolog of BRCA1 that is associated with familial ovarian cancer predisposition in humans, develop benign epithelial tumors in their reproductive tract. These tumors are driven, at least in part, by a prolongation of the proestrus phase of the estrus cycle (equivalent to the follicular phase of the menstrual cycle) in Brca1 mutant mice, resulting in prolonged unopposed estrogen stimulation. Mutant mice synchronized in proestrus also showed increased circulating estradiol levels, but the possibility that this change also has a role in tumor predisposition was not investigated. We sought to determine whether these changes in hormonal stimulation result in measurable changes in tissues targeted by estrogen outside the ovary. Here we show that mice carrying a Brca1 mutation in their ovarian granulosa cells show increased endometrial proliferation during proestrus, implying that the effects of Brca1 inactivation on estrogen stimulation have short-term consequences, at least on this target organ. We further show that mutant mice develop increased femoral trabecular thickness and femoral length, which are well-known consequences of chronic estrogen stimulation. Estrogen biosynthesis by granulosa cells was increased not only in mice carrying a homozygous Brca1 mutation, but also in heterozygous mutants mimicking the mutational status in granulosa cells of human BRCA1 mutation carriers. The results suggest that human germline BRCA1 mutations, although associated with increased cancer risk, may also have beneficial consequences, such as increased bone strength, that may have contributed to the maintenance of mutated BRCA1 alleles in the human gene pool.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Animals, Newborn
  • Aromatase / metabolism
  • BRCA1 Protein / genetics
  • BRCA1 Protein / metabolism*
  • Cell Differentiation
  • Cell Proliferation
  • Endometrium / growth & development*
  • Endometrium / metabolism
  • Estradiol / blood
  • Estrogens / biosynthesis
  • Estrous Cycle / blood
  • Estrous Cycle / genetics*
  • Estrus Synchronization / physiology
  • Female
  • Femur / metabolism
  • Femur / pathology
  • Genes, BRCA1
  • Genetic Predisposition to Disease
  • Genotype
  • Granulosa Cells / metabolism*
  • Granulosa Cells / pathology
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Osteoclasts / cytology
  • Osteoclasts / drug effects
  • Osteoclasts / metabolism
  • Stem Cells / cytology
  • Stem Cells / drug effects

Substances

  • BRCA1 Protein
  • Estrogens
  • Estradiol
  • Aromatase