Changes in androgen receptor nongenotropic signaling correlate with transition of LNCaP cells to androgen independence

E Unni, S Sun, B Nan, MJ McPhaul, B Cheskis… - Cancer research, 2004 - AACR
E Unni, S Sun, B Nan, MJ McPhaul, B Cheskis, MA Mancini, M Marcelli
Cancer research, 2004AACR
A cure for prostate cancer (CaP) will be possible only after a complete understanding of the
mechanisms causing this disease to progress from androgen dependence to androgen
independence. To carry on a careful characterization of the phenotypes of CaP cell lines
before and after acquisition of androgen independence, we used two human CaP LNCaP
sublines: LNCaPnan, which is androgen dependent (AD), and LNCaP-HP, which is
androgen independent (AI). In AD LNCaPnan cells, dihydrotestosterone (DHT) stimulated in …
Abstract
A cure for prostate cancer (CaP) will be possible only after a complete understanding of the mechanisms causing this disease to progress from androgen dependence to androgen independence. To carry on a careful characterization of the phenotypes of CaP cell lines before and after acquisition of androgen independence, we used two human CaP LNCaP sublines: LNCaPnan, which is androgen dependent (AD), and LNCaP-HP, which is androgen independent (AI). In AD LNCaPnan cells, dihydrotestosterone (DHT) stimulated in an androgen receptor (AR)-dependent way a phosphorylation signaling pathway involving steroid receptor coactivator (Src)–mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase (MEK)-1/2–ERK-1/2–cAMP-response element binding-protein (CREB). Activation of this pathway was associated with increased [3H]thymidine incorporation and resistance to apoptosis. Use of dominant-negative forms of MEK-1/2 and CREB demonstrated in LNCaPnan cells that DHT induced [3H]thymidiine incorporation through a thus far unidentified molecule activated downstream of MEK-1/2, and antiapoptosis through phosphorylation of the transcription factor CREB. In contrast, in AI LNCaP-HP cells, the Src–MEK-1/2–ERK-1/2–CREB pathway was constitutively active. Because it was not further stimulated by addition of DHT, no increase of [3H]thymidine incorporation or apoptosis resistance was demonstrated in LNCaP-HP cells. Additional experiments showed that Src and the scaffold protein MNAR coimmunoprecipitated with AR, indicating a role for Src as an apical molecule in the Src–MEK-1/2–ERK-1/2–CREB pathway. Interestingly, differences between the two cell lines were that in LNCaP-HP cells presence of an AI phenotype and lack of response to DHT were associated with constitutive activation of the protein kinase Src and interaction among Src, AR, and MNAR. In contrast, in LNCaPnan cells, presence of an AD phenotype and ability to respond to DHT were associated with DHT-dependent activation of Src kinase activity and interaction among Src, AR, and MNAR. Intriguingly, in LNCaPnan cells, we found that transcription through the prototypical CREB-responsive promoter c-fos could be induced in a DHT-dependent way, and this action was inhibited by the AR antagonist Casodex and MEK-1 inhibitor PD98059. In contrast, transcription through the PSA P/E promoter, a prototypical AR-dependent promoter directly activated by agonist, was obliterated only by Casodex. Additional experiments with genital skin fibroblasts derived from patients with a variety of AR abnormalities indicated that nongenotropic AR signaling does not depend on an intact DNA-binding domain or on the ability of AR to translocate to the nucleus. The results suggest the following: (1) Constitutive activation of the Src–MEK-1/2–ERK-1/2–CREB pathway is associated with the AI phenotype observed in LNCaP-HP cells. (2) Activation of the Src–MEK-1/2–ERK-1/2–CREB pathway is DHT dependent in AD LNCaPnan cells. (3) DHT activation of this pathway is associated with induction of [3H]thymidine incorporation by a molecule activated downstream of MEK-1/2 and of antiapoptosis through activation of the transcription factor CREB in AD LNCaPnan cells. (4) AR regulates transcription either directly upon ligand binding and nuclear translocation or indirectly through kinase pathways leading to activation of downstream transcription factors. (5) Nuclear translocation and ability of the DNA-binding domain of AR to interact with DNA are not prerequisites for nongenotropic AR activity.
AACR