An Estrogen Receptor-α Knock-In Mutation Provides Evidence of Ligand-Independent Signaling and Allows Modulation of Ligand-Induced Pathways in Vivo

KW Sinkevicius, JE Burdette, K Woloszyn…�- …, 2008 - academic.oup.com
KW Sinkevicius, JE Burdette, K Woloszyn, SC Hewitt, K Hamilton, SL Sugg, KA Temple…
Endocrinology, 2008academic.oup.com
Estrogen-nonresponsive estrogen receptor-α (ERα) knock-in (ENERKI) mice were
generated to distinguish between ligand-induced and ligand-independent ER-α actions in
vivo. These mice have a mutation [glycine 525 to leucine (G525L)] in the ligand-binding
domain of ERα, which significantly reduces ERα interaction with and response to
endogenous estrogens, whereas not affecting growth factor activation of ligand-independent
pathways. ENERKI mice had hypoplastic uterine tissues and rudimentary mammary gland�…
Estrogen-nonresponsive estrogen receptor-α (ERα) knock-in (ENERKI) mice were generated to distinguish between ligand-induced and ligand-independent ER-α actions in vivo. These mice have a mutation [glycine 525 to leucine (G525L)] in the ligand-binding domain of ERα, which significantly reduces ERα interaction with and response to endogenous estrogens, whereas not affecting growth factor activation of ligand-independent pathways. ENERKI mice had hypoplastic uterine tissues and rudimentary mammary gland ductal trees. Females were infertile due to anovulation, and their ovaries contained hemorrhagic cystic follicles because of chronically elevated levels of LH. The ENERKI phenotype confirmed that ligand-induced activation of ERα is crucial in the female reproductive tract and mammary gland development. Growth factor treatments induced uterine epithelial proliferation in ovariectomized ENERKI females, directly demonstrating that ERα ligand-independent pathways were active. In addition, the synthetic ERα selective agonist propyl pyrazole triol (PPT) and ER agonist diethylstilbestrol (DES) were still able to activate ligand-induced G525L ERα pathways in vitro. PPT treatments initiated at puberty stimulated ENERKI uterine development, whereas neonatal treatments were needed to restore mammary gland ductal elongation, indicating that neonatal ligand-induced ERα activation may prime mammary ducts to become more responsive to estrogens in adult tissues. This is a useful model for in vivo evaluation of ligand-induced ERα pathways and temporal patterns of response. DES did not stimulate an ENERKI uterotrophic response. Because ERβ may modulate ERα activation and have an antiproliferative function in the uterus, we hypothesize that ENERKI animals were particularly sensitive to DES-induced inhibition of ERα due to up-regulated uterine ERβ levels.
Oxford University Press