Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 154(7): 2446-56, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23696568

ABSTRACT

Tumor growth is fostered by inhibition of cell death, which involves the receptiveness of tumor to growth factors and hormones. We have recently shown that testosterone exerts proapoptotic effects in prostate and colon cancer cells through a membrane-initiated mechanism. In addition, we have recently reported that dehydroepiandrosterone (DHEA) can control cell fate, activating nerve growth factor (NGF) receptors, namely tropomyosin-related kinase (Trk)A and p75 neurotrophin receptor, in primary neurons and in PC12 tumoral cells. NGF was recently involved in cancer cell proliferation and apoptosis. In the present study, we explored the cross talk between androgens (testosterone and DHEA) and NGF in regulating apoptosis of prostate and colon cancer cells. DHEA and NGF strongly blunted serum deprivation-induced apoptosis, whereas testosterone induced apoptosis of both cancer cell lines. The antiapoptotic effect of both DHEA and NGF was completely reversed by testosterone. In line with this, DHEA or NGF up-regulated, whereas testosterone down-regulated, the expression of TrkA receptor. The effects of androgens were abolished in both cell lines in the presence of TrkA inhibitor. DHEA induced the phosphorylation of TrkA and the interaction of p75 neurotrophin receptor with its effectors, Rho protein GDP dissociation inhibitor and receptor interacting serine/threonine-protein kinase 2. Conversely, testosterone was unable to activate both receptors. Testosterone acted as a DHEA and NGF antagonist, by blocking the activation of both receptors by DHEA or NGF. Our findings suggest that androgens may influence hormone-sensitive tumor cells via their cross talk with NGF receptors. The interplay between steroid hormone and neurotrophins signaling in hormone-dependent tumors offers new insights in the pathophysiology of these neoplasias.


Subject(s)
Dehydroepiandrosterone/pharmacology , Receptors, Nerve Growth Factor/metabolism , Testosterone/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Humans , Male , Nerve Growth Factor/pharmacology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , PC12 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Rats , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
2.
FEBS J ; 279(7): 1231-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22309306

ABSTRACT

The serum- and glucocorticoid-dependent kinases 1-3 (SGK1-3) are downstream effectors of phosphatidylinositol 3-kinases, implicated in various cell responses including colon cancer tumorigenesis in mice. Here, we investigated the role of SGK1 in the regulation of cell motility. Using Caco-2 colon tumor and HEK293 embryonic kidney cells, we report that transfection with the constitutively active SGK1 mutant (SGK1-SD) significantly enhanced cell motility. The cell-adhesion protein vinculin was effectively dephosphorylated in SGK1-SD-transfected cells. Treatment of the cells with phosphatase inhibitors restored vinculin phosphorylation and inhibited cell migration, indicating a significant role for vinculin phosphorylation in SGK1-induced motility. SGK1-SD-enhanced cell motility was inhibited by activation of membrane androgen-binding sites (mAR) via testosterone-conjugates in both cell lines, whereas intracellular androgen receptor (iAR)-silencing and flutamide treatment revealed that these effects were clearly independent of the interaction of SGK1 with the classical androgen receptors (iAR). More importantly, mAR activation restored vinculin phosphorylation in SGK1-SD-transfected cells, whereas silencing of vinculin fully reversed the mAR-induced inhibition of the migratory capacity, implying that this protein is directly involved in cell motility regulation by SGK1 and mAR. This study indicates for the first time that SGK1 regulates cell migration via vinculin dephosphorylation, a mechanism that is controlled by mAR function.


Subject(s)
Cell Movement/physiology , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Androgen/metabolism , Vinculin/metabolism , Animals , Caco-2 Cells , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Receptors, Androgen/genetics , Vinculin/genetics
3.
Mol Cancer ; 7: 88, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-19055752

ABSTRACT

BACKGROUND: Recently we have reported membrane androgen receptors-induced apoptotic regression of prostate cancer cells regulated by Rho/ROCK/actin signaling. In the present study we explored the specificity of these receptors and we analyzed downstream effectors controlling survival and apoptosis in hormone refractory DU145-prostate cancer cells stimulated with membrane androgen receptor-selective agonists. RESULTS: Using membrane impermeable conjugates of serum albumin covalently linked to testosterone, we show here down-regulation of the activity of pro-survival gene products, namely PI-3K/Akt and NF-kappaB, in DU145 cells. Testosterone-albumin conjugates further induced FasL expression. A FasL blocking peptide abrogated membrane androgen receptors-dependent apoptosis. In addition, testosterone-albumin conjugates increased caspase-3 and Bad protein activity. The actin cytoskeleton drug cytochalasin B and the ROCK inhibitor Y-27632 inhibited FasL induction and caspase-3 activation, indicating that the newly identified Rho/Rock/actin signaling may regulate the downstream pro-apoptotic effectors in DU145 cells. Finally, other steroids or steroid-albumin conjugates did not interfere with these receptors indicating testosterone specificity. CONCLUSION: Collectively, our results provide novel mechanistic insights pointing to specific pro-apoptotic molecules controlling membrane androgen receptors-induced apoptotic regression of prostate cancer cells and corroborate previously published observations on the potential use of membrane androgen receptor-agonists as novel anti-tumor agents in prostate cancer.


Subject(s)
Androgens , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/chemistry , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Fas Ligand Protein/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serum Albumin/chemistry , Serum Albumin/pharmacology , Signal Transduction/drug effects , Testosterone/chemistry , Testosterone/pharmacology , bcl-Associated Death Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...