Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Cancers (Basel) ; 11(12)2019 Dec 12.
Article En | MEDLINE | ID: mdl-31842254

Dysfunctions of androgen/TGF-ß signaling play important roles in prostate tumorigenesis. Prostate Transmembrane Protein Androgen Induced 1 (PMEPA1) inhibits androgen and TGF-ß signaling via a negative feedback loop. The loss of PMEPA1 confers resistance to androgen signaling inhibitors and promotes bone metastasis. Conflicting reports on the expression and biological functions of PMEPA1 in prostate and other cancers propelled us to investigate isoform specific functions in prostate cancer (PCa). One hundred and twenty laser capture micro-dissection matched normal prostate and prostate tumor tissues were analyzed for correlations between quantitative expression of PMEPA1 isoforms and clinical outcomes with Q-RT-PCR, and further validated with a The Cancer Genome Atlas (TCGA) RNA-Seq dataset of 499 PCa. Cell proliferation was assessed with cell counting, plating efficiency and soft agar assay in androgen responsive LNCaP and TGF-ß responsive PC3 cells. TGF-ß signaling was measured by SMAD dual-luciferase reporter assay. Higher PMEPA1-a mRNA levels indicated biochemical recurrence (p = 0.0183) and lower PMEPA1-b expression associated with metastasis (p = 0.0173). Further, lower PMEPA1-b and a higher ratio of PMEPA1-a vs. -b were correlated to higher Gleason scores and lower progression free survival rate (p < 0.01). TGF-ß-responsive PMEPA1-a promoted PCa cell growth, and androgen-responsive PMEPA1-b inhibited cancer cell proliferation. PMEPA1 isoforms -a and -b were shown to be promising candidate biomarkers indicating PCa aggressiveness including earlier biochemical relapse and lower disease specific life expectancy via interrupting androgen/TGF-ß signaling.

3.
Mol Cancer Res ; 15(10): 1308-1317, 2017 10.
Article En | MEDLINE | ID: mdl-28607007

The oncogenic activation of the ETS-related gene (ERG) due to gene fusions is present in over half of prostate cancers in Western countries. Because of its high incidence and oncogenic role, ERG and components of ERG network have emerged as potential drug targets for prostate cancer. Utilizing gene expression datasets, from matched normal and prostate tumor epithelial cells, an association of NOTCH transcription factors with ERG expression status was identified, confirming that NOTCH factors are direct transcriptional targets of ERG. Inhibition of ERG in TMPRSS2-ERG-positive VCaP cells led to decreased levels of NOTCH1 and 2 proteins and downstream transcriptional targets and partially recapitulated the phenotypes associated with ERG inhibition. Regulation of NOTCH1 and 2 genes by ERG were also noted with ectopic ERG expression in LNCaP (ERG-negative prostate cancer) and RWPE-1 (benign prostate-derived immortalized) cells. Furthermore, inhibition of NOTCH by the small-molecule γ-secretase inhibitor 1, GSI-1, conferred an increased sensitivity to androgen receptor (AR) inhibitors (bicalutamide and enzalutamide) or the androgen biosynthesis inhibitor (abiraterone) in VCaP cells. Combined treatment with bicalutamide and GSI-1 showed strongest inhibition of AR, ERG, NOTCH1, NOTCH2, and PSA protein levels along with decreased cell growth, cell survival, and enhanced apoptosis. Intriguingly, this effect was not observed in ERG-negative prostate cancer cells or immortalized benign/normal prostate epithelial cells. These data underscore the synergy of AR and NOTCH inhibitors in reducing the growth of ERG-positive prostate cancer cells.Implications: Combinational targeting of NOTCH and AR signaling has therapeutic potential in advanced ERG-driven prostate cancers. Mol Cancer Res; 15(10); 1308-17. ©2017 AACR.


Androgen Antagonists/pharmacology , Oligopeptides/pharmacology , Prostatic Neoplasms/genetics , Receptors, Notch/genetics , Androstenes/pharmacology , Anilides/pharmacology , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Humans , Male , Nitriles/pharmacology , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Notch/metabolism , Tosyl Compounds/pharmacology , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
4.
Sci Rep ; 7(1): 1109, 2017 04 24.
Article En | MEDLINE | ID: mdl-28439080

Mechanistic studies of deregulated ERG in prostate cancer and other cancers continue to enhance its role in cancer biology and its utility as a biomarker and therapeutic target. Here, we show that ERG, through its physical interaction with androgen receptor, induces AR aggregation and endoplasmic reticulum stress in the prostate glands of ERG transgenic mice. Histomorphological alterations and the expression of ER stress sensors Atf6, Ire1α, Perk, their downstream effectors Grp78/BiP and eIF2α in ERG transgenic mouse prostate glands indicate the presence of chronic ER stress. Transient activation of apoptotic cell death during early age correlated well with the differential regulation of ER stress sensors, in particular Perk. Epithelial cells derived from ERG transgenic mouse prostates have increased prostasphere formation with resistance to radiation induced cell death. Continued activation of cell survival factors, Atf6 and Ire1α during chronic ER stress due to presence of ERG in prostate epithelium induces survival pathways and provides a selection pressure in the continuum of ERG dependent neoplastic process. These novel insights will enhance the understanding of the mechanistic functions of ERG in prostate tumor biology and towards development of early targeted therapeutic strategies for prostate cancer.


Endoplasmic Reticulum Stress , Prostatic Neoplasms/physiopathology , Protein Aggregation, Pathological , Receptors, Androgen/metabolism , Animals , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Histocytochemistry , Immunohistochemistry , Male , Mice, Transgenic , Microscopy , Prostate/pathology , Transcriptional Regulator ERG/metabolism
5.
Oncotarget ; 7(16): 22791-806, 2016 Apr 19.
Article En | MEDLINE | ID: mdl-26988912

Epigenetic regulation by SIRT1, a multifaceted NAD+-dependent protein deacetylase, is one of the most common factors modulating cellular processes in a broad range of diseases, including prostate cancer (CaP). SIRT1 is over-expressed in CaP cells, however the associated mechanism is not well understood. To identify whether specific microRNAs might mediate this linkage, we have screened a miRNA library for differential expression in ERG-associated CaP tissues. Of 20 differentially and significantly expressed miRNAs that distinguish ERG-positive tumors from ERG-negative tumors, we find miR-449a is highly suppressed in ERG-positive tumors. We establish that SIRT1 is a direct target of miR-449a and is also induced by ERG in ERG-associated CaP. Our data suggest that attenuation of miR-449a promotes the invasive phenotype of the ERG-positive CaP in part by inducing the expression of SIRT1 in prostate cancer cells. Furthermore, we also find that suppression of SIRT1 results in a significant reduction in ERG expression in ERG-positive CaP cells, indicating a feed-back regulatory loop associated with ERG, miR-449a and SIRT1. We also report that ERG suppresses p53 acetylation perhaps through miR-449a-SIRT1 axis in CaP cells. Our findings provide new insight into the function of miRNAs in regulating ERG-associated CaP. Thus, miR-449a activation or SIRT1 suppression may represent new therapeutic opportunity for ERG-associated CaP.


Gene Expression Regulation, Neoplastic/physiology , MicroRNAs/metabolism , Prostatic Neoplasms/pathology , Sirtuin 1/biosynthesis , Cell Line, Tumor , Humans , Male , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Phenotype , Prostatic Neoplasms/genetics , Transcriptional Regulator ERG/genetics
6.
Int J Cancer ; 133(1): 31-42, 2013 Jul.
Article En | MEDLINE | ID: mdl-23280553

TNFAIP8 is a NF-κB-inducible, oncogenic molecule. Previous "promoter array" studies have identified differential methylation and regulation of TNFAIP8 in prostate epithelial and cancer cell lines. Here we demonstrate that TNFAIP8 expression is induced by androgen in hormone-responsive LNCaP prostate cancer cells. In athymic mice bearing hormone-refractory PC-3 prostate tumor xenografts, intravenous treatment with a liposomal formulation of TNFAIP8 antisense oligonucleotide (LE-AS5) caused reduced expression of TNFAIP8 in tumor tissues, and a combination of LE-AS5 and radiation or docetaxel treatment resulted in significant inhibition of PC-3 tumor growth as compared to single agents. The immunohistochemical evaluation of TNFAIP8 expression revealed correlation of both cytoplasmic and nuclear TNFAIP8 overexpression with high grade prostatic adenocarcinomas, while nuclear overexpression was found to be an independent predictor of disease recurrence controlling for tumor grade. Increased nuclear TNFAIP8 expression was statistically significantly associated with a 2.44 fold (95 % confidence interval: 1.01-5.91) higher risk of prostate cancer recurrence. Mechanistically, TNFAIP8 seems to function as a scaffold (or adaptor) protein. In the antibody microarray analysis of proteins associated with the TNFAIP8 immune-complex, we have identified Karyopherin alpha2 as a novel binding partner of nuclear TNFAIP8 in PC-3 cells. The Ingenuity Pathway Analysis of the TNFAIP8 interacting proteins suggested that TNFAIP8 influences cancer progression pathways and networks involving integrins and matrix metalloproteinases. Taken together, present studies demonstrate that TNFAIP8 is a novel therapeutic target in prostate cancer, and indicate a potential relationship of the nuclear trafficking of TNFAIP8 with adverse outcomes in a subset of prostate cancer patients.


Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/metabolism , Oligonucleotides, Antisense/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Taxoids/therapeutic use , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Chemotherapy, Adjuvant , Disease Progression , Docetaxel , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunohistochemistry , Liposomes , Male , Mice , Mice, Nude , Neoplasm Grading , Oligonucleotides, Antisense/chemical synthesis , Predictive Value of Tests , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Array Analysis , Radiotherapy, Adjuvant , Transplantation, Heterologous , Up-Regulation
7.
J Carcinog ; 10: 37, 2011.
Article En | MEDLINE | ID: mdl-22279422

Prevalent gene fusions involving regulatory sequences of the androgen receptor (AR) regulated genes (primarily TMPRSS2) and protein coding sequences of nuclear transcription factors of the ETS gene family (predominantly ERG) result in unscheduled androgen dependent ERG expression in prostate cancer (CaP).Cumulative data from a large number of studies in the past six years accentuate ERG alterations in more than half of all CaP patients in Western countries. Studies underscore that ERG functions are involved in the biology of CaP. ERG expression in normal context is selective to endothelial cells, specific hematopoetic cells and pre-cartilage cells. Normal functions of ERG are highlighted in hematopoetic stem cells. Emerging data continues to unravel molecular and cellular mechanisms by which ERG may contribute to CaP. Herein, we focus on biological and clinical aspects of ERG oncogenic alterations, potential of ERG-based stratification of CaP and the possibilities of targeting the ERG network in developing new therapeutic strategies for the disease.

8.
J Cancer ; 1: 197-208, 2010 Oct 25.
Article En | MEDLINE | ID: mdl-21060730

Oncogenic activation of the ETS Related Gene (ERG) in humans was originally identified in subsets of Ewing sarcomas, myeloid leukemias and, recently, in the majority of prostate cancers. Expression of human ERG protein and consequently its functions in normal and disease states needs to be better understood in light of its suggested role in cell differentiation and proliferation. Here, we analyzed temporal and spatial expression of the Erg (mouse protein) by immunohistochemical analysis during mouse embryonic and adult organogenesis using a highly specific ERG monoclonal antibody (ERG MAb). This study establishes widespread immunolocalization of Erg protein in endothelial cells and restricted expression in precartilage and hematopoietic tissues. Intriguingly, Erg is not expressed in any epithelial tissue including prostate epithelium, or in infiltrating lymphocytes that are occasionally seen in the prostate environment, a common site of tumors with ERG rearrangements and unscheduled ERG expression. These findings will further aid in investigations of Erg functions in normal and disease conditions.

9.
Matrix Biol ; 28(3): 129-36, 2009 Apr.
Article En | MEDLINE | ID: mdl-19379665

Targeted disruption of the dentin sialophosphoprotein (DSPP) gene in the mice (Dspp(-/-)) results in dentin mineralization defects with enlarged predentin phenotype similar to human dentinogenesis imperfecta type III. Using DSPP/biglycan (Dspp(-/-)Bgn(-/0)) and DSPP/decorin (Dspp(-/-)Dcn(-/-)) double knockout mice, here we determined that the enlarged predentin layer in Dspp(-/-) teeth is rescued in the absence of decorin, but not in the absence of biglycan. However, Fourier transform infrared (FTIR) spectroscopy analysis reveals similar hypomineralization of dentin in both Dspp(-/-)Bgn(-/0) and Dspp(-/-)Dcn(-/-) teeth. Atomic force microscopy (AFM) analysis of collagen fibrils in dentin shows subtle differences in the collagen fibril morphology in these genotypes. The reduction of enlarged predentin in Dspp(-/-)Dcn(-/-) mice suggests that the elevated level of decorin in Dspp(-/-) predentin interferes with the mineralization process at the dentin mineralization front. On the other hand, the lack of DSPP and biglycan leads to the increased number of calcospherites in Dspp(-/-)Bgn(-/0) predentin, suggesting that a failure in coalescence of calcospherites was augmented in Dspp(-/-)Bgn(-/0) teeth as compared to Dspp(-/-) teeth. These findings indicate that normal expression of small leucine rich proteoglycans, such as biglycan and decorin, plays an important role in the highly orchestrated process of dentin mineralization.


Dentin/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Tooth Calcification/genetics , Animals , Biglycan , Collagen/ultrastructure , Decorin , Dentin/diagnostic imaging , Dentin/pathology , Dentin/ultrastructure , Dentinogenesis Imperfecta/genetics , Dentinogenesis Imperfecta/pathology , Humans , Incisor/diagnostic imaging , Incisor/growth & development , Incisor/metabolism , Incisor/pathology , Mice , Mice, Inbred Strains , Mice, Knockout , Microradiography , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Odontoblasts/metabolism , Phosphoproteins , Protein Precursors/genetics , Sialoglycoproteins , Spectroscopy, Fourier Transform Infrared
10.
Eur J Oral Sci ; 114 Suppl 1: 30-4; discussion 39-41, 379, 2006 May.
Article En | MEDLINE | ID: mdl-16674659

Transforming growth factor-beta1 (TGF-beta1) is a key regulator of many cellular processes, including cell adhesion, the immune response and synthesis of extracellular matrix proteins. In the present study, we report the characterization of enamel defects in a transgenic mouse model overexpressing TGF-beta1 in odontoblasts and ameloblasts, its expression being driven by the promoter sequences of the dentin sialophosphoprotein gene. As reported earlier, these mice develop distinct dentin defects similar to those seen in human dentin dysplasia and dentinogenesis imperfecta. A further detailed examination of enamel in these mice revealed that from the early secretory stage, ameloblasts began to detach from dentin to form cyst-like structures. A soft X-ray analysis revealed that this cyst-like structure had a disorganized and partially mineralized matrix with an abnormal mineralization pattern and a globular appearance. In the molars, the enamel was not only pitted and hypoplastic, but enamel rods were completely lost. Thus, altered TGF-beta1 expression in the tooth seems to trigger detachment of ameloblasts and abnormal secretion and deposition of minerals in the cyst-like structures adjoining the dentin. We speculate that the altered expression of TGF-beta1 in teeth impacts the adhesion process of ameloblasts to dentin.


Ameloblasts/ultrastructure , Dental Enamel/abnormalities , Transforming Growth Factor beta/genetics , Ameloblasts/metabolism , Animals , Cell Adhesion/genetics , Dental Enamel/ultrastructure , Dental Enamel Hypoplasia/genetics , Dental Enamel Hypoplasia/pathology , Dentin/abnormalities , Dentin/ultrastructure , Dentin Dysplasia/genetics , Dentin Dysplasia/pathology , Dentinogenesis Imperfecta/genetics , Dentinogenesis Imperfecta/pathology , Extracellular Matrix Proteins , Gene Expression Regulation/genetics , Mice , Mice, Transgenic , Microradiography , Microscopy, Electron, Scanning , Odontoblasts/ultrastructure , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Protein Precursors/genetics , Sialoglycoproteins/genetics , Tooth Calcification/genetics , Transforming Growth Factor beta1
...