Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 105(13): 5236-41, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18356297

ABSTRACT

Androgen receptor (AR) mediates transcriptional activation of diverse target genes through interactions with various coactivators that may alter its function and help mediate the switch between prostate cell proliferation and differentiation. We recently identified p44/MEP50 as an AR coactivator and further showed that it is expressed primarily in the nucleus and cytoplasm of benign prostate epithelial and prostate cancer cells, respectively. We also showed that haploinsufficiency in p44(+/-) mice causes prostate epithelial cell proliferation. To establish direct cause-and-effect relationships, we have used p44 fusion proteins that are selectively expressed in the nucleus or cytoplasm of prostate cancer cells (LNCaP), along with RNAi analyses, to examine effects of p44 both in vitro and in vivo (in tumor xenografts). We show that preferential expression of p44 in the nucleus inhibits proliferation of LNCaP cells in an AR-dependent manner, whereas preferential expression of p44 in the cytoplasm enhances cell proliferation. These effects appear to be mediated, at least in part, through the regulation of distinct cell-cycle regulatory genes that include p21 (up-regulated by nuclear p44) and cyclin D2 and CDK6 (up-regulated by cytoplasmic p44). Importantly, we also demonstrate that altered p44 expression is associated with androgen-independent prostate cancer. Our results indicate that nuclear p44 and cytoplasmic p44 have distinct and opposing functions in the regulation of prostate cancer cell proliferation.


Subject(s)
Cytoplasm/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Androgens/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Separation , Cyclin D2 , Cyclin-Dependent Kinase 6/metabolism , Cyclins/metabolism , Enzyme Activation , Humans , Male , Receptors, Androgen/metabolism , Transcription Factors/genetics
2.
J Cell Mol Med ; 12(6B): 2790-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18266956

ABSTRACT

Androgen receptor (AR) is expressed in both stromal and epithelial cells of the prostate. The majority of studies on AR expression and function in prostate cancer is focused on malignant epithelial cells rather than stromal cells. In this study, we examined the levels of stromal AR in androgen-dependent and -independent prostate cancer and the function of stromal AR in prostate cancer growth and invasion. We showed that stromal AR levels were decreased in the areas surrounding cancerous tissue, especially in androgen-independent cancer. Using two telomerase-immortalized human stromal cell lines, one AR-positive and the other AR-negative, we demonstrated that stromal cells lacking AR stimulated cell proliferation of co-cultured prostate cancer cells in vitro and enhanced tumour growth in vivo when co-injected with PC3 epithelial cells in nude mice. In contrast, stromal cells expressing AR suppressed prostate cancer growth in vitro and in vivo. In parallel with cancer growth, in vitro invasion assays revealed that stromal cells lacking AR increased the invasion ability of PC3 cell by one order of magnitude, while stromal cells expressing AR reduced this effect. These results indicate a negative regulation of prostate cancer growth and invasion by stromal AR. This provides potentially new mechanistic insights into the failure of androgen ablation therapy, and the reactivation of stromal AR could be a novel therapeutic approach for treating hormone refractory prostate cancer.


Subject(s)
Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Stromal Cells/metabolism , Aged , Androgens/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/ultrastructure , Stromal Cells/pathology , Stromal Cells/ultrastructure , Telomerase/metabolism
3.
Am J Pathol ; 172(1): 225-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18156210

ABSTRACT

ARA70 was first identified as a gene fused to the ret oncogene in thyroid carcinoma and subsequently as a co-activator for androgen receptor (AR). Two isoforms of ARA70 have been identified: a 70-kDa version called ARA70 alpha and an internally spliced 35-kDa variant termed ARA70 beta. We have previously reported that ARA70 alpha expression is reduced in prostate cancer, and its overexpression inhibits proliferation of LNCaP prostate cancer cells. However, the function of the ARA70 beta isoform in prostate cancer is not understood. In this report we examined the effects of ARA70 beta on AR transcriptional regulation as well as prostate cancer cellular proliferation and invasion. Although both ARA70 alpha and ARA70 beta functioned as transcriptional co-activators of AR in cell-based reporter assays, ARA70 beta overexpression, in contrast to ARA70 alpha, promoted prostate cancer cellular proliferation and invasion through Matrigel. Interestingly, genome-wide expression profiling of cells expressing ARA70 beta revealed an increase in the expression of genes involved in the control of cell division and adhesion, compatible with a role for ARA70 beta in proliferation and invasion. Consistent with its function in promoting cell growth and invasion, ARA70 beta expression was increased in prostate cancer. Our findings implicate ARA70 beta as a regulator of tumor cell growth and metastasis by affecting gene expression.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Oncogene Proteins/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , Collagen/chemistry , Drug Combinations , Humans , Laminin/chemistry , Male , Models, Biological , Neoplasm Invasiveness , Nuclear Receptor Coactivators , Prostatic Neoplasms/metabolism , Proteoglycans/chemistry , Retroviridae/metabolism , Transcription, Genetic , Transfection
4.
J Mol Endocrinol ; 36(3): 463-83, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16720717

ABSTRACT

Androgen receptor (AR) is a ligand-activated transcription factor that mediates the action of androgens and is essential for the growth, function, and cell differentiation of the prostate gland. Here, we demonstrated that the prostate apoptosis response factor-4 (par-4) functions as a novel AR coactivator. Par-4 physically interacted with the DNA-binding domain of AR, enhanced AR interaction with DNA, and increased AR-dependent transcription. Par-4 enhanced the c-FLIP promoter activity and was recruited on to the c-FLIP gene in the presence of androgens, and the dominant-negative par-4 decreased c-FLIP expression. These results suggest that, in addition to its proapoptotic function, par-4 acts as a novel transcription cofactor for AR to target c-FLIP gene expression. In addition, we demonstrated that loss of c-FLIP expression was essential for castration-induced apoptosis in the prostate gland and that enhanced c-FLIP expression was associated with prostate cancer progression to the androgen-resistant stage. Our data shed light on a transcription-mediated mechanism for the effects of the AR pathway on cell survival and apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Survival/physiology , Gene Expression Regulation , Prostate/physiology , Receptors, Androgen/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Castration , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Peptides/genetics , Peptides/metabolism , Prostate/cytology , Prostate/metabolism , Protein Structure, Quaternary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Elements, Transcriptional , Transcription, Genetic
5.
Mol Biol Cell ; 14(11): 4592-604, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12960422

ABSTRACT

We evaluated the hypothesis that the N-terminal region of the replication control protein Cdc6 acts as an inhibitor of cyclin-dependent kinase (Cdk) activity, promoting mitotic exit. Cdc6 accumulation is restricted to the period from mid-cell cycle until the succeeding G1, due to proteolytic control that requires the Cdc6 N-terminal region. During late mitosis, Cdc6 is present at levels comparable with Sic1 and binds specifically to the mitotic cyclin Clb2. Moderate overexpression of Cdc6 promotes viability of CLB2Deltadb strains, which otherwise arrest at mitotic exit, and rescue is dependent on the N-terminal putative Cdk-inhibitory domain. These observations support the potential for Cdc6 to inhibit Clb2-Cdk, thus promoting mitotic exit. Consistent with this idea, we observed a cytokinesis defect in cdh1Delta sic1Delta cdc6Delta2-49 triple mutants. However, we were able to construct viable strains, in three different backgrounds, containing neither SIC1 nor the Cdc6 Cdk-inhibitory domain, in contradiction to previous work. We conclude, therefore, that although both Cdc6 and Sic1 have the potential to facilitate mitotic exit by inhibiting Clb2-Cdk, mitotic exit nevertheless does not require any identified stoichiometric inhibitor of Cdk activity.


Subject(s)
Cell Cycle Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle , Cell Cycle Proteins/genetics , Cell Survival/genetics , Cell Survival/physiology , Cyclin B , Cyclin-Dependent Kinase Inhibitor Proteins , Mass Spectrometry , Mitosis , Protein Binding , Protein Structure, Tertiary/genetics , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...