Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Signal ; 27(11): 2261-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26257301

ABSTRACT

While androgen deprivation therapy (ADT) reduces tumor burden, autocrine growth factor loops such as human epidermal growth factor receptor 2 (HER2/ErbB-2/neu) have been proposed to contribute to prostate cancer (PCa) survival and relapse. However, the role of ErbB-2 in regulating androgen-sensitive (AS) and castration-resistant (CR) cell proliferation remains unclear. Here, we determined the role of ErbB-2 in PCa progression and survival under steroid-reduced conditions using two independent PCa cell progression models. In AR-positive androgen-independent (AI) PCa cells that exhibit the CR phenotype, ErbB-2 was constitutively activated, compared to corresponding AS PCa cells. In AS LNCaP C-33 cells, androgen-induced ErbB-2 activation through ERK1/2 mediates PCa cell proliferation. Further, the ErbB-2-specific but not EGFR-specific inhibitor suppresses basal and androgen-stimulated cell proliferation and also blocks ERK1/2 activation. ErbB-2 ectopic expression and cPAcP siRNA transfection of LNCaP C-33 cells each increases ErbB-2 tyrosine phosphorylation, correlating with increased AI PSA secretion and cell proliferation. Conversely, trapping ErbB-2 by transfected endoplasmic reticulum-targeting ScFv5R expression vector abolished DHT-induced LNCaP C-33 cell growth. Moreover, inhibition of ErbB-2 but not EGFR in AI LNCaP C-81 and MDA PCa2b-AI PCa cells significantly abolished AI cell growth. In contrast to androgens via ErbB-2/ERK1/2 signaling in AS PCa cells, the inhibition of ErbB-2 abrogated AI cell proliferation by inhibiting the cell survival protein Akt in those AI cells. These results suggest that ErbB-2 is a prominent player in mediating the ligand-dependent and -independent activation of AR in AS and AI/CR PCa cells respectively for PCa progression and survival.


Subject(s)
Androgen Antagonists/therapeutic use , ErbB Receptors/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptor, ErbB-2/metabolism , Receptors, Androgen/metabolism , Acid Phosphatase/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Endoplasmic Reticulum , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kallikreins/metabolism , Male , Phosphorylation/genetics , Prostate-Specific Antigen/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Signal Transduction
2.
J Biol Chem ; 285(31): 23598-606, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20498373

ABSTRACT

Cellular prostatic acid phosphatase (cPAcP), an authentic tyrosine phosphatase, is proposed to function as a negative growth regulator of prostate cancer (PCa) cells in part through its dephosphorylation of ErbB-2. Nevertheless, the direct interaction between cPAcP and ErbB-2 has not been shown nor the specific dephosphorylation site of ErbB-2 by cPAcP. In this report, our data show that the phosphorylation level of ErbB-2 primarily at Tyr(1221/2) correlates with the growth rate of both LNCaP and MDA PCa2b human PCa cells. Further, cPAcP reciprocally co-immunoprecipitated with ErbB-2 in a non-permissive growth condition. Expression of wild type cPAcP, but not inactive mutant, by cDNA in cPAcP-null LNCaP C-81 cells results in decreased tyrosine phosphorylation of ErbB-2 including Tyr(1221/2). Concurrently, Tyr(317) phosphorylation of p52(Shc), proliferating cell nuclear antigen expression, and cell growth are decreased in these cells. Conversely, decreased cPAcP expression by short hairpin RNA in LNCaP C-33 cells was associated with elevated phosphorylation of ErbB-2 initially at Tyr(1221/2). Its downstream p52(Shc), ERK1/2, Akt, Src, STAT-3, and STAT-5 were activated, and cell proliferation, proliferating cell nuclear antigen, and cyclin D1 expression were increased. Stable subclones of C-33 cells by small interfering PAcP had elevated Tyr(1221/2) phosphorylation of ErbB-2 and exhibited androgen-independent growth and increased tumorigenicity in xenograft female animals. In summary, our data together indicate that in prostate epithelia, cPAcP interacts with and dephosphorylates ErbB-2 primarily at Tyr(1221/2) and hence blocks downstream signaling, leading to reduced cell growth. In PCa cells, decreased cPAcP expression is associated with androgen-independent cell proliferation and tumorigenicity as seen in advanced hormone-refractory prostate carcinomas.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Protein Tyrosine Phosphatases/physiology , Receptor, ErbB-2/metabolism , Acid Phosphatase , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Mice , Neoplasm Transplantation , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Subcellular Fractions , Tyrosine/chemistry
3.
Prostate ; 67(5): 557-71, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17221842

ABSTRACT

BACKGROUND: The expression of prostate-derived factor (PDF) is significantly elevated in human prostate tumors. We investigate the functional role and signaling of PDF in androgen receptor (AR)-positive human prostate cancer cells. METHODS: Transient or stable expression of PDF by cDNA transfection, antisense-mediated gene silencing, media conditioned by PDF-elevated cells, and antibody (Ab) neutralization were employed. RESULTS: Elevated endogenous and exogenous expression of PDF and treatment of PDF-enriched media were associated with increased proliferation and clonogenic growth of the cells. On the contrary, knockdown of PDF or addition of PDF neutralizing Ab resulted in diminished proliferation and reduced anchorage-independent growth. Further, ERK1/2 and p90RSK, but not Smad2/3, were activated in PDF-elevated cells as well as in cells treated with PDF-enriched media, while inhibition of ERK1/2 decreased the growth of those cells. CONCLUSION: PDF promotes AR-positive prostate tumor progression through upregulating cell proliferation via ERK1/2 signal pathway.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Blotting, Western , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Silencing , Growth Differentiation Factor 15 , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Transfection , Transforming Growth Factor beta/metabolism
4.
Endocr Relat Cancer ; 12(4): 805-22, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322323

ABSTRACT

Human prostatic acid phosphatase (PAcP) was used as a valuable surrogate marker for monitoring prostate cancer prior to the availability of prostate-specific antigen (PSA). Even though the level of PAcP is increased in the circulation of prostate cancer patients, its intracellular level and activity are greatly diminished in prostate cancer cells. Recent advances in understanding the function of the cellular form of PAcP (cPAcP) have shed some light on its role in prostate carcinogenesis, which may have potential applications for prostate cancer therapy. It is now evident that cPAcP functions as a neutral protein tyrosine phosphatase (PTP) in prostate cancer cells and dephosphorylates HER-2/ErbB-2/Neu (HER-2: human epidermal growth factor receptor-2) at the phosphotyrosine (p-Tyr) residues. Dephosphorylation of HER-2 at its p-Tyr residues results in the down-regulation of its specific activity, which leads to decreases in growth and tumorigenicity of those cancer cells. Conversely, decreased cPAcP expression correlates with hyperphosphorylation of HER-2 at tyrosine residues and activation of downstream extracellular signal-regulated kinase (ERK)/mitogen activated protein kinase (MAPK) signaling, which results in prostate cancer progression as well as androgen-independent growth of prostate cancer cells. These in vitro results on the effect of cPAcP on androgen-independent growth of prostate cancer cells corroborate the clinical findings that cPAcP level is greatly decreased in advanced prostate cancer and provide insights into one of the molecular mechanisms involved in prostate cancer progression. Results from experiments using xenograft animal models further indicate a novel role of cPAcP as a tumor suppressor. Future studies are warranted to clarify the use of cPAcP as a therapeutic agent in human prostate cancer patients.


Subject(s)
Androgens/metabolism , Prostatic Neoplasms/enzymology , Protein Tyrosine Phosphatases/metabolism , Receptor, ErbB-2/metabolism , Tumor Suppressor Proteins/metabolism , Acid Phosphatase , Cell Proliferation , Epithelium/enzymology , Humans , Male , Prostatic Neoplasms/drug therapy , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/therapeutic use , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/therapeutic use
5.
Int J Cancer ; 108(5): 672-8, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14696093

ABSTRACT

Members of Shc family conventionally serve as critical adaptors in tyrosine phosphorylation signal transduction pathways. p66(Shc) protein, a member of Shc family, is predominantly expressed in epithelial cells, whereas the regulation of its expression remains an enigma. We describe the effect of steroid hormones on the protein level of p66(Shc) and growth stimulation in hormone-sensitive human prostate, testicular and breast cancer cells. In DHT-treated androgen-sensitive prostate cancer LNCaP C-33 cells, the protein level of p66(Shc) was elevated by approximately 3-fold, correlating with increased cell growth. This DHT effect on p66(Shc) protein level and growth regulation was also observed in another androgen-sensitive prostate cancer cell line MDA PCa2b as well as 2 testicular cancer cell lines, Tera-1 and Tera-2 cells. Similarly, the female sex hormone estrogen had a stimulating effect on p66(Shc) protein level and proliferation in estrogen-sensitive MCF-7 breast cancer cells. The upregulation of p66(Shc) protein level by DHT was competitively abolished by Casodex, an androgen antagonist used to treat prostate cancer. Moreover, immunohistochemical analyses showed that the p66(Shc) protein level was significantly higher in primary prostate tumors than in adjacent non-cancerous cells (p < 0.05). The data collectively indicate that p66(Shc) protein levels correlate with steroid hormone-stimulated cell growth and prostate carcinogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/biosynthesis , Dihydrotestosterone/pharmacology , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Division , Estrogens/pharmacology , Humans , Male , Phosphorylation , Prostate/metabolism , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Testicular Neoplasms/metabolism , Tumor Cells, Cultured , Up-Regulation
6.
Biochem Biophys Res Commun ; 305(3): 598-604, 2003 Jun 06.
Article in English | MEDLINE | ID: mdl-12763036

ABSTRACT

As a part of the study to identify genes associated with hormone-refractory stage of human prostate cancer, we have recently identified several genetic and epigenetic changes that seem to be associated with the progression of androgen-sensitive to androgen-independent prostate tumor cells. In the present study, we report a novel gene, macrophage inhibitory cytokine-1 (MIC-1) also known as prostate derived factor (PDF), that was highly expressed in androgen-independent LNCaP-C81 cells and its metastatic variant LNCaP-Ln3 compared to androgen-sensitive LNCaP-C33 cells. The MIC-1/PDF expression was dysregulated (very low to non-detectable) in the androgen-independent PC3 and DU145 cells. Interestingly, serum factors demonstrated a differential regulation of MIC-1/PDF in the androgen-sensitive and the androgen-independent cells of LNCaP cells. Immunohistochemical analysis on 15 prostatic adenocarcinomas showed a weak staining in the benign prostatic glandular area (intensity score 2.38+/-0.25; n=13), while the immunoreactivity was significantly stronger (p<0.05) in areas of adenocarcinoma (score 7.33+/-0.88; n=15). Altogether, these data suggest that the serum factors (including androgens and cytokines) might contribute to the regulation of the MIC-1/PDF gene that seems to be associated with the progression of prostate cancer.


Subject(s)
Adenocarcinoma/metabolism , Cytokines/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Bone Morphogenetic Proteins , Cell Culture Techniques , Cytokines/analysis , Cytokines/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Growth Differentiation Factor 15 , Humans , Immunohistochemistry , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
7.
J Immunol ; 168(10): 4846-53, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11994433

ABSTRACT

Decoy receptor 3 (DcR3), a soluble receptor belonging to the TNFR superfamily, is a receptor for both Fas ligand (FasL) and LIGHT. It has been demonstrated that DcR3 is up-regulated in lung and colon cancers, thus promoting tumor growth by neutralizing the cytotoxic effects of FasL and LIGHT. In this study, we found that DcR3.Fc profoundly modulated dendritic cell differentiation and maturation from CD14(+) monocytes, including the up-regulation of CD86/B7.2, and the down-regulation of CD40, CD54/ICAM-1, CD80/B7.1, CD1a, and HLA-DR. Moreover, DcR3-treated dendritic cells suppressed CD4(+) T cell proliferation in an allogeneic MLR and up-regulated IL-4 secretion of CD4(+)CD45RA(+) T cells. This suggests that DcR3.Fc may act not only as a decoy receptor to FasL and LIGHT, but also as an effector molecule to skew T cell response to the Th2 phenotype.


Subject(s)
Adjuvants, Immunologic/physiology , Dendritic Cells/cytology , Dendritic Cells/immunology , Receptors, Cell Surface/physiology , Adjuvants, Immunologic/metabolism , Animals , COS Cells , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/metabolism , Fas Ligand Protein , HT29 Cells , Humans , Jurkat Cells , Ligands , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Member 6b , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14 , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...