Your browser does not support JavaScript!

Home    Μη γενομική δράση στεροειδών ορμονών : μοριακοί μηχανισμοί ρύθμισης και βιολογικός ρόλος στη νεοπλασία  

Results - Details

Add to Basket
[Add to Basket]
Identifier 000347667
Title Μη γενομική δράση στεροειδών ορμονών : μοριακοί μηχανισμοί ρύθμισης και βιολογικός ρόλος στη νεοπλασία
Author Παπαδοπούλου, Ναταλία
Thesis advisor Στουρνάρας, Χρήστος
Reviewer Γραβάνης, Αχιλλέας
Καρδάσης, Δημήτρης
Παπαματθαιάκης, Σ.
Θεοδωρόπουλος, Παναγιώτης
Τσατσάνης, Χρήστος
Παπακωνσταντή, Ε.
Abstract Prostate cancer is a leading cause of premature death in Western countries. After many decades, androgen-ablation remains the principal treatment paradigm for PCa. However, relapse from hormonal therapy is almost certain and most patients with metastasis progress to end-stage disease regardless of treatment strategy. The lack of progress on clinical options for PCa patients reflects our poor understanding of the molecular and cellular mechanisms that underlie disease etiology and progression. The non-genomic action of steroids is an active field of research during the last decade. Whereas the classical (genomic) model of steroid action involves binding to specific intracellular steroid receptors, translocation to the nucleus, DNA binding, and activation of specific genes, a process which requires hours to be completed, a nongenomic effect usually occurs within minutes, is insensitive to inhibitors of transcription and translation and it may occur in cells not expressing or expressing non-functional classical steroid receptors. Recently, membrane androgen receptors (mAR’s) have been detected in the human prostate cancer cell lines LNCaP and DU145, the second of which is devoid of functional classical intracellular receptors (iAR’s). 􀈂􀇹R activation through nonpermeable androgen analogues like testosterone-BSA induces the apoptotic regression of both prostate cancer cell lines in vitro and in vivo. Moreover, a new, non-genomic mechanism of androgen action was found to alter actin cytoskeleton dynamics in LNCaP cells through a signaling cascade involving phosphorylation and activation of FAK, PI-3K and the downstream signaling molecules Rac1 and Cdc42. Actin cytoskeleton reorganization is one of the earliest cellular responses to many extracellular stimuli. Recently it was reported to control apoptosis in different cellular systems. Based on these findings the main goal of the present study was to examine a possible link between rapid actin cytoskeleton reorganization and the apoptotic response of prostate cancer cells following mAR activation. DU145 cells expressing nonfunctional intracellular androgen receptors (iAR’s) were chosen as the principal cellular model for this study, in order to exclude interference of genomic androgen action in the examined cellular responses. In line with previous results in LNCaP cells, both dihydrotestosterone and the nonpermeable analogue testosterone-BSA, induced rapid actin polymerization followed by the formation of filopodia and stress fibers in DU145 cells, as indicated by the 19 decrease of the G/total actin ratio and morphological analysis using confocal laser scanning microscopy. Furthermore, mAR activation led to actin microfilament redistribution and apoptosis in PC3 cells, another prostate cancer cell line expressing non-functional iAR’s. The mAR induced apoptotic response of all three cell lines was inhibited by pre-incubation of the cells with actin microfilament blocking agents such as cytochalasin B (cytB) or phallacidin, indicating a key role for actin cytoskeleton dynamics in regulating apoptosis in mAR stimulated prostate cancer cells. We then addressed the signaling pathway downstream of mAR activation in DU145 cells and identified a novel, non-genomic signaling cascade leading to actin cytoskeleton reorganization, different from the one described in LNCaP cells. Indeed, the levels of phosphorylated FAK were found to be comparatively high in nonstimulated DU145 cells, probably due to the cell’s high invasive and metastatic potential. Cell stimulation by testosterone-BSA did not induce further phosphorylation of FAK or its immediate substrate PI-3K. The levels of endogenous phospho-PI-3K instead decreased after prolonged incubation with the androgen, while co-treatment of cells with the PI-3K inhibitor wortmannin did not affect androgen induced actin reorganization. In addition, mAR activation had no effect on Rac1-GTP levels in DU145 cells. These findings indicate that the FAK 􀁯 PI-3K 􀁯 Rac1 pathway is not involved in regulating actin cytoskeleton dynamics in DU145 cells. On the contrary, three other members of the family of small RhoGTPases, namely RhoA, RhoB and Cdc42, are rapidly activated following mAR stimulation in DU145 cells. Cells expressing dominant-negative RhoA, RhoB or Cdc42 failed to form new actin stress fibers and filopodia in response to testosterone-BSA, whereas the surrounding non-transfected cells showed robust actin reorganization. Furthermore, ROCK was found to be their downstream effector, since inhibition of ROCK by the specific inhibitor Y-27632 completely blocked actin polymerization. RhoGTPases are known to regulate the activity of LIM kinases. We therefore performed in vitro kinase assays and detected rapid testosterone-BSA induced LIMK2 phosphorylation. Destrin, an isoform of cofilin, co-precipitated with LIMK2 and became phosphorylated with kinetics similar to those of LIMK2. Since the members of the ADF/cofilin family have an established role as actin depolymerizing factors, phosphorylation and hence deactivation of destrin contributes to increased actin polymerization in our system. In addition, ROCK inhibition by Y-27632 completely blocked androgen induced 20 LIMK2/destrin phosphorylation. These findings suggest that mAR activation leads to actin cytoskeleton reorganization through a RhoA/B, Cdc42 􀁯 ROCK 􀁯 LIMK2 􀁯 destrin signaling cascade in androgen-independent DU145 cells. Interestingly, RhoA and RhoB were activated in response to testosterone-BSA in androgen-dependent LNCaP cells too, though their activation required longer incubation times with the androgen. Treatment with the specific ROCK inhibitor, Y- 27632, also blocked actin polymerization in LNCaP cells. Furthermore, the androgen induced apoptotic response of both DU145 and LNCaP cells was abolished upon treatment with Y-27632. On the contrary cell treatment with the PI-3K inhibitor, wortmannin, decreased apoptotic rates in LNCaP cells exclusively, probably by blocking the signal transmission to the actin cytoskeleton upstream of Rho/ROCK. Therefore, it seems that the Rho/ROCK pathway regulates mAR induced apoptosis by controlling actin cytoskeleton reorganization in prostate cancer cells, bearing or not functional intracellular androgen receptors (iAR's). Finally, we studied the pro-apoptotic factors involved in the apoptotic response of DU145 cells. The expression of members of the Bcl-2 protein family was not effected, but testosterone-BSA treatment resulted in rapid activation of caspase-3 and increased FasL expression. Both factors are controlled by Rho/ROCK and actin cytoskeleton dynamics, as indicated by the abolishment of their activation in the presence of the specific ROCK inhibitor, Y-27632, or actin blocking agents. In addition, the rapid decrease of Akt phosphorylation in response to testosterone-BSA, with kinetics similar to those of PI-3K, might suggest the deactivation of the main survival pathway PI-3K/Akt, thereby contributing to apoptosis. In conclusion, the present study has identified, for the first time, the biological significance of the mAR induced rapid actin cytoskeleton reorganization in human prostate cancer cells, demonstrating a key role for the actin cytoskeleton in regulating the cell's apoptotic response. In addition, a new, non-genomic signaling pathway leading to androgen induced actin polymerization in DU145 prostate cancer cells, expressing non-functional iAR's, was identified. This pathway involves the activation of RhoGTPases and of their downstream kinases ROCK and LIMK2, transmitting a signal to destrin, thereby modulating its ability to shift the actin polymerization equilibrium in a positive manner. Interestingly, Rho/ROCK were also shown to 21 control actin cytoskeleton reorganization in iAR-positive LNCaP cells. Furthermore, the regulatory role of Rho/ROCK in the apoptotic response of both iAR- positive LNCaP and iAR-negative DU145 cells has been highlighted. Finally, experimental data indicating the rapid activation of pro-apoptotic factors in response to mAR stimulation, such as the increase in caspase-3 activity and the deactivation of the PI- 3K/Akt survival pathway, provide a starting point for a mechanistical approach of the interaction between the actin cytoskeleton and apoptosis in prostate cancer cells.
Language Greek
Subject Prostatic Neoplasms
Steroids
Προστάτη νεοπλάσματα
Στεροειδή
Issue date 2007-07-14
Collection   School/Department--School of Medicine--Department of Medicine--Doctoral theses
  Type of Work--Doctoral theses
Views 315

Digital Documents
No preview available

Download document
View document
Views : 2