po 058 unravelling the protective role of androgens androgen receptorin breast cancer when bad goes good

Abstract

Introduction Androgen receptor (AR) expression in breast cancer growth and progression appears to be clinically relevant and disease context specific. In oestrogen receptor (ER)α-positive primary breast cancers, AR positivity correlates with lower tumour grade and a better clinical outcome. These clinical-pathological findings mirror the capability of androgens to counteract ERα-dependent proliferation in both normal mammary epithelium and ERα-positive breast cancer preclinical models in which androgen/AR-dependent pro-apoptotic effects have been also evidenced. Here we report a novel additional mechanism underlining the protective, anti-proliferative role exerted by AR signalling. This mechanism involves modulation of the expression, cellular distribution and function of BAD, a pro-apoptotic member of the Bcl-2 family proteins, whose expression is related to a significantly better disease free survival in (ER)α-positive human breast cancers. Material and methods MCF-7, TD47D, ZR-75 breast cancer cells; qReal Time PCR; western blotting (WB); immunofluorescence analysis (IF); immunoprecipitation assay (IP); DNA affinity precipitation assay; Chromatin Immunoprecipitation Assay. Results and discussions The expression of a panel of pro/anti-apoptotic proteins was investigated in cellular protein lysates from ERα/AR-positive MCF-7 cells cultured for 1, 3 and 6 days under androgen treatment. The expression of the anti-apoptotic Bcl-2 protein, or the pro-apoptotic BID and BAX remained unchanged, while a sustained increase in the expression of the pro-apoptotic BAD could be observed, reducing the Bcl-2/BAD ratio and, thus, shifting the delicate balance between inhibitors and inducers of cell death. Interestingly, androgens induced a marked BAD levels increase into the nuclear compartment in ERα/AR-positive MCF-7, T47D and ZR-75 as well as in ERα negative/AR-positive SKBR3 cells. The androgen-regulated intracellular localization of BAD involved an AR/BAD physical interaction, suggesting a nuclear role for BAD upon androgen stimulation. Indeed, androgens induced both AR and BAD recruitment at a AP-1 and at a ARE site within the cyclin D1 promoter region, contributing to explain the anti-proliferative effect of androgens in breast cancer cells. Conclusion We defined a novel mechanism by which androgens modulate BAD expression and force its ability to act as a cell cycle inhibitor through modulation of cyclin D1 gene transcriptional activity, strengthening the protective role of androgen signalling in estrogen-responsive breast cancer

    Similar works