Enhancing the effectiveness of androgen deprivation in prostate cancer by inducing Filamin A nuclear localization

    1. Paramita M Ghosh1,2
    1. 1VA Northern California Health Care System, Mather, California, USA
      2Department of Urology, University of California Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, California 95817, USA
      3University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
    1. (Correspondence should be addressed to P M Ghosh at Department of Urology, University of California Davis School of Medicine; Email: paramita.ghosh{at}ucdmc.ucdavis.edu)

    Abstract

    As prostate cancer (CaP) is regulated by androgen receptor (AR) activity, metastatic CaP is treated with androgen deprivation therapy (ADT). Despite initial response, patients on ADT eventually progress to castration-resistant CaP (CRPC), which is currently incurable. We previously showed that cleavage of the 280 kDa structural protein Filamin A (FlnA) to a 90 kDa fragment, and nuclear localization of the cleaved product, sensitized CRPC cells to ADT. Hence, treatment promoting FlnA nuclear localization would enhance androgen responsiveness. Here, we show that FlnA nuclear localization induced apoptosis in CRPC cells during ADT, identifying it as a treatment tool in advanced CaP. Significantly, the natural product genistein combined polysaccharide (GCP) had a similar effect. Investigation of the mechanism of GCP-induced apoptosis showed that GCP induced FlnA cleavage and nuclear localization and that apoptosis resulting from GCP treatment was mediated by FlnA nuclear localization. Two main components of GCP are genistein and daidzein: the ability of GCP to induce G2 arrest was due to genistein whereas sensitivity to ADT stemmed from daidzein; hence, both were needed to mediate GCP's effects. FlnA cleavage is regulated by its phosphorylation; we show that ADT enhanced FlnA phosphorylation, which prevented its cleavage, whereas GCP inhibited FlnA phosphorylation, thereby sensitizing CaP cells to ADT. In a mouse model of CaP recurrence, GCP, but not vehicle, impeded relapse following castration, indicating that GCP, when administered with ADT, interrupted the development of CRPC. These results demonstrate the efficacy of GCP in promoting FlnA nuclear localization and enhancing androgen responsiveness in CaP.

    • Revision received 30 August 2012
    • Accepted 18 September 2012
    • Made available online as an Accepted Preprint 19 September 2012
    | Table of Contents