Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer ; 15(1): 68, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27809841

ABSTRACT

BACKGROUND: The CXCL12/CXCR4 axis transactivates HER2 and promotes intraosseous tumor growth. To further explore the transactivation of HER2 by CXCL12, we investigated the role of small GTP protein Gαi2 in Src and HER2 phosphorylation in lipid raft membrane microdomains and the significance of CXCR4 in prostate cancer bone tumor growth. METHODS: We used a variety of methods such as lipid raft isolation, invasion assays, an in vivo model of intratibial tumor growth, bone histomorphometry, and immunohistochemistry to determine the role of CXCR4 signaling in lipid raft membrane microdomains and effects of targeting of CXCR4 for bone tumor growth. RESULTS: We determined that (a) CXCL12/CXCR4 transactivation of EGFR and HER2 is confined to lipid raft membrane microdomains, (b) CXCL12 activation of HER2 and Src is mediated by small GTP proteins in lipid rafts, (c) inhibition of the CXCL12/CXCR4 axis through plerixafor abrogates the initial establishment of tumor growth without affecting the growth of established bone tumors, and (d) inhibition of EGFR signaling through gefitinib leads to inhibition of established bone tumor growth. CONCLUSIONS: These data suggest that lipid raft membrane microdomains are key sites for CXCL12/CXCR4 transactivation of HER2 via small GTP binding protein Gαi2 and Src kinase. The initial establishment of prostate cancer is supported by the endosteal niche, and blocking the CXCL12/CXCR4 axis of this niche along with its downstream signaling severely compromises initial establishment of tumors in the bone microenvironment, whereas expanding bone tumors are sensitive only to the members of growth factor receptor inhibition.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Chemokine CXCL12/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Animals , Benzylamines , Bone Neoplasms/drug therapy , Cell Line, Tumor , Cyclams , Disease Models, Animal , ErbB Receptors/agonists , ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heterocyclic Compounds/pharmacology , Humans , Male , Membrane Microdomains/metabolism , Mice , Phosphorylation , Prostatic Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
2.
Clin Cancer Res ; 21(11): 2569-79, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25754347

ABSTRACT

PURPOSE: Intratumoral androgen synthesis in prostate cancer contributes to the development of castration-resistant prostate cancer (CRPC). Several enzymes responsible for androgen biosynthesis have been shown to be overexpressed in CRPC, thus contributing to CRPC in a castrated environment. The TMPRSS2-ERG transcription factor has been shown to be present in primary prostate cancer tumors as well as CRPC tumors. We hypothesize that TMPRSS2-ERG fusions regulate androgen biosynthetic enzyme (ABE) gene expression and the production of androgens, which contributes to the development of CRPC. EXPERIMENTAL DESIGN: We used a panel of assays, including lentivirus transduction, gene expression, chromatin immunoprecipitation and sequencing, liquid chromatography-mass spectrometric quantitation, immunocytochemistry, immunohistochemistry, and bioinformatics analysis of gene microarray databases, to determine ERG regulation of androgen synthesis. RESULTS: We found that ERG regulated the expression of the ABE AKR1C3 in prostate cancer cells via direct binding to the AKR1C3 gene. Knockdown of ERG resulted in reduced AKR1C3 expression, which caused a reduction in both DHT synthesis and PSA expression in VCaP prostate cancer cells treated with 5α-androstanedione (5α-Adione), a DHT precursor metabolite. Immunohistochemical staining revealed that ERG was coexpressed with AKR1C3 in prostate cancer tissue samples. CONCLUSIONS: These data suggest that AKR1C3 catalyzes the biochemical reduction of 5α-Adione to DHT in prostate cancer cells, and that ERG regulates this step through upregulation of AKR1C3 expression. Elucidation of ERG regulation of ABEs in CRPC may help to stratify TMPRSS2-ERG fusion-positive prostate cancer patients in the clinic for anti-androgen receptor-driven therapies; and AKR1C3 may serve as a valuable therapeutic target in the treatment of CRPC.


Subject(s)
3-Hydroxysteroid Dehydrogenases/biosynthesis , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Prostatic Neoplasms, Castration-Resistant/genetics , Serine Endopeptidases/genetics , Trans-Activators/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3 , Androgen Antagonists/administration & dosage , Androgens/biosynthesis , Androgens/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Male , Oncogene Proteins, Fusion/genetics , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Signal Transduction/drug effects , Trans-Activators/biosynthesis , Transcriptional Regulator ERG
3.
Mol Cancer Res ; 11(11): 1349-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23918819

ABSTRACT

UNLABELLED: CXCR4 is a chemokine receptor that mediates invasion and metastasis. CXCR4 expression is transcriptionally regulated in cancer cells and is associated with aggressive prostate cancer phenotypes. Previously, we and others have shown that the transcription factor ERG regulates CXCR4 expression in prostate cancer cells and that androgens modulate CXCR4 expression via increasing ERG expression. Herein, the molecular mechanisms of ERG-mediated CXCR4 promoter activation, phosphorylation of ERG by intracellular kinases and subsequent CXCR4 expression, as well as the status of ERG and CXCR4 in human prostate cancer specimens were investigated. Using multiple molecular strategies, it was demonstrated that (i) ERG expressed in TMPRSS2-ERG fusion positive VCaP cells selectively binds to specific ERG/Ets bindings sites in the CXCR4 promoter; (ii) distal binding sites mediate promoter activation; (iii) exogenously expressed ERG promotes CXCR4 expression; (iv) ERG is phosphorylated at Serine-81 and -215, by both IKK and Akt kinases, and Akt mediates CXCR4 expression; (v) ERG-induced CXCR4 drives CXCL12-dependent adhesion to fibronectin; and (vi) ERG and CXCR4 were coexpressed in human prostate cancer tissue, consistent with ERG-mediated transcriptional activation of CXCR4. These data demonstrate that ERG activates CXCR4 expression by binding to specific ERG/Ets responsive elements and via intracellular kinases that phosphorylate ERG at discrete serine residues. IMPLICATIONS: These findings provide a mechanistic link between TMPRSS2-ERG translocations and intracellular kinase-mediated phosphorylation of ERG on enhanced metastasis of tumor cells via CXCR4 expression and function in prostate cancer cells.


Subject(s)
Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prostatic Neoplasms/genetics , Receptors, CXCR4/metabolism , Trans-Activators/metabolism , Binding Sites/genetics , Cell Adhesion/physiology , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Neoplasm Invasiveness , Phosphorylation , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Receptors, CXCR4/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction/genetics , Trans-Activators/genetics , Transcriptional Regulator ERG , Tumor Microenvironment
4.
Mol Cancer ; 12(1): 85, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902739

ABSTRACT

INTRODUCTION: The chemokine CXCL12, also known as SDF-1, and its receptor, CXCR4, are overexpressed in prostate cancers and in animal models of prostate-specific PTEN deletion, but their regulation is poorly understood. Loss of the tumor suppressor PTEN (phosphatase and tensin homolog) is frequently observed in cancer, resulting in the deregulation of cell survival, growth, and proliferation. We hypothesize that loss of PTEN and subsequent activation of Akt, frequent occurrences in prostate cancer, regulate the CXCL12/CXCR4 signaling axis in tumor growth and bone metastasis. METHODS: Murine prostate epithelial cells from PTEN+/+, PTEN+/-, and PTEN-/- (prostate specific knockdown) mice as well as human prostate cancer cell lines C4-2B, PC3, and DU145 were used in gene expression and invasion studies with Akt inhibition. Additionally, HA-tagged Akt1 was overexpressed in DU145, and tumor growth in subcutaneous and intra-tibia bone metastasis models were analyzed. RESULTS: Loss of PTEN resulted in increased expression of CXCR4 and CXCL12 and Akt inhibition reversed expression and cellular invasion. These results suggest that loss of PTEN may play a key role in the regulation of this chemokine activity in prostate cancer. Overexpression of Akt1 in DU145 resulted in increased CXCR4 expression, as well as increased proliferation and cell cycle progression. Subcutaneous injection of these cells also resulted in increased tumor growth as compared to neo controls. Akt1 overexpression reversed the osteosclerotic phenotype associated with DU145 cells to an osteolytic phenotype and enhanced intra-osseous tumor growth. CONCLUSIONS: These results suggest the basis for activation of CXCL12 signaling through CXCR4 in prostate cancer driven by the loss of PTEN and subsequent activation of Akt. Akt1-associated CXCL12/CXCR4 signaling promotes tumor growth, suggesting that Akt inhibitors may potentially be employed as anticancer agents to target expansion of PC bone metastases.


Subject(s)
Chemokine CXCL12/metabolism , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL12/genetics , Humans , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Receptors, CXCR4/genetics , Signal Transduction
5.
J Cancer Sci Ther ; 4(4): 94-101, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-23264855

ABSTRACT

TMPRSS2-Ets gene fusions were identified in prostate cancers where the promoter of transmembrane protease, serine 2 (TMPRSS2) fused with coding sequence of the erythroblastosis virus E26 (Ets) gene family members. TMPRSS2 is an androgen responsive transmembrane serine protease. Ets family members are oncogenic transcription factors that contain a highly conserved Ets DNA binding domain and an N-terminal regulatory domain.Fusion of these gene results in androgen dependent transcription of Ets factor in prostate tumor cells. The ERG is the most common fusion partner with TMPRSS2 promoter in prostate cancer patients. The high prevalence of these gene fusions, in particular TMPRSS2-ERG, makes them attractive as potential diagnostic and prognostic indicators, as well as making them a potential target for tailored therapies.This review focuses on the clinical and biological significance of TMPRSS2-ERG fusions and their role in PC development and progression.

6.
Am J Pathol ; 180(3): 1017-1027, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22209699

ABSTRACT

Platelet-derived growth factor (PDGF) family members are potent growth factors that regulate cell proliferation, migration, and transformation. Clinical studies have shown that both PDGF receptor ß (ß-PDGFR) and its ligand PDGF D are up-regulated in primary prostate cancers and bone metastases, whereas PDGF B, a classic ligand for ß-PDGFR, is not frequently detected in clinical samples. In this study, we examined the role of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN) in the regulation of PDGF expression levels using both a prostate-specific, conditional PTEN-knockout mouse model and mouse prostate epithelial cell lines established from these mice. We found an increase in PDGF D and ß-PDGFR expression levels in PTEN-null tumor cells, accompanied by a decrease in PDGF B expression. Among Akt isoforms, increased Akt3 expression was most prominent in mouse PTEN-null cells, and phosphatidylinositol 3-kinase/Akt activity was essential for the maintenance of increased PDGF D and ß-PDGFR expression. In vitro deletion of PTEN resulted in a PDGF ligand switch from PDGF B to PDGF D in normal mouse prostate epithelial cells, further demonstrating that PTEN regulates this ligand switch. Similar associations between PTEN status and PDGF isoforms were noted in human prostate cancer cell lines. Taken together, these results suggest a mechanism by which loss of PTEN may promote prostate cancer progression via PDGF D/ß-PDGFR signal transduction.


Subject(s)
PTEN Phosphohydrolase/physiology , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/physiology , Animals , Humans , Ligands , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/physiology , Tumor Cells, Cultured , Up-Regulation
7.
Prostate ; 72(12): 1328-38, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22213159

ABSTRACT

BACKGROUND: The major cause of death in prostate cancer (PCa) cases is due to distant metastatic lesions, with the bone being the most prevalent site for secondary colonization. Utilization of small molecule inhibitors to treat bone metastatic PCa have had limited success either as monotherapies or in combination with other chemotherapeutics due to intolerable toxicities. In the current study, we developed a clinically relevant in vivo intraosseous tumor model overexpressing the platelet-derived growth factor D (PDGF D) to test the efficacy of a newly characterized vascular endothelial growth factor receptor (VEGFR)/PDGFR inhibitor, cediranib (also called AZD2171). METHODS: An intratibial-injection model was established utilizing DU145 cells with or without increased PDGF D expression. Tumor-bearing mice were treated by daily gavage administration of cediranib and/or weekly i.p. injection of docetaxel for 7 weeks. Tibiae were monitored by in vivo/ex vivo X-rays and histomorphometry analysis was performed to estimate tumor volume and tumor-associated trabecular bone growth. RESULTS: Cediranib reduced intraosseous growth of prostate tumors as well as tumor-associated bone responses. When compared to the standard chemotherapeutic agent docetaxel, cediranib exhibited a stronger inhibition of tumor-associated bone response. The efficacy of cediranib was further enhanced when the drug was co-administered with docetaxel. Importantly, the therapeutic benefits of cediranib and docetaxel are more prominent in intraosseous prostate tumors overexpressing PDGF D. CONCLUSION: These novel findings support the utilization of cediranib, either alone or in combination with docetaxel, to treat bone metastatic PCa exhibiting PDGF D expression.


Subject(s)
Bone Neoplasms/drug therapy , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Growth Inhibitors/therapeutic use , Lymphokines/biosynthesis , Platelet-Derived Growth Factor/biosynthesis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Quinazolines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Humans , Lymphokines/antagonists & inhibitors , Male , Mice , Mice, SCID , Platelet-Derived Growth Factor/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Random Allocation , Xenograft Model Antitumor Assays/methods
8.
Cancer Res ; 70(23): 9631-40, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21098708

ABSTRACT

Increasing evidence indicates the significance of platelet-derived growth factor receptor-ß (ß-PDGFR) signaling in prostate cancer (PCa). Accordingly, preclinical studies suggest the potential of ß-PDGFR as a therapeutic target in metastatic PCa. However, a ligand responsible for ß-PDGFR activation in PCa was unknown, and recent clinical trials with imatinib mesylate showed limited success due to normal tissue toxicity. Similarly, in spite of mounting evidence indicating the significance of matriptase in PCa, little is known about its substrates or molecular actions during PCa progression. Here, we identified PDGF-D as a ligand for ß-PDGFR in PCa and discovered matriptase as its regulator. Matriptase activates PDGF-D by proteolytic removal of the CUB domain in a 2-step process, creating a hemidimer, followed by growth factor domain dimer (GFD-D) generation. Matriptase can deactivate PDGF-D by further proteolytic cleavage within the GFD, revealing its biphasic regulation. Importantly, PDGF-D/matriptase colocalization is accompanied with ß-PDGFR phosphorylation in human PCa tissues. This study unveiled a novel signaling axis of matriptase/PDGF-D/ß-PDGFR in PCa, providing new insights into functional interplay between serine protease and growth factor signaling networks.


Subject(s)
Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Serine Endopeptidases/metabolism , Signal Transduction , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Humans , In Situ Hybridization , Lymphokines/chemistry , Lymphokines/genetics , Male , Mice , Microscopy, Confocal , Mutation , NIH 3T3 Cells , Phosphorylation , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , Protein Multimerization , RNA Interference , Receptor, Platelet-Derived Growth Factor beta/genetics , Serine Endopeptidases/genetics
9.
Biochim Biophys Acta ; 1803(11): 1287-97, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20620173

ABSTRACT

Membrane type 1 (MT1)-matrix metalloproteinase (MT1-MMP) is a membrane-tethered MMP that has been shown to play a key role in promoting cancer cell invasion. MT1-MMP is highly expressed in bone metastasis of prostate cancer (PC) patients and promotes intraosseous tumor growth of PC cells in mice. The majority of metastatic prostate cancers harbor loss-of-function mutations or deletions of the tumor suppressor PTEN (phosphatase and tensin homologue deleted on chromosome ten). However, the role of PTEN inactivation in MT1-MMP expression in PC cells has not been examined. In this study, prostate epithelial cell lines derived from mice that are either heterozygous (PTEN(+/-)) or homozygous (PTEN(-/-)) for PTEN deletion or harboring a wild-type PTEN (PTEN(+/+)) were used to investigate the expression of MT1-MMP. We found that biallelic loss of PTEN is associated with posttranslational regulation of MT1-MMP protein in mouse PC cells. PTEN(-/-) PC cells display higher levels of MT1-MMP at the cell surface when compared to PTEN(+/+) and PTEN(+/-) cells and consequently exhibited enhanced migratory and collagen-invasive activities. MT1-MMP displayed by PTEN(-/-) cells is differentially O-glycosylated and exhibits a slow rate of turnover. MT1-MMP expression in PTEN(-/-) cells is under control of the PI3K/AKT signaling pathway, as determined using pharmacological inhibitors. Interestingly, rapamycin, an mTOR inhibitor, upregulates MT1-MMP expression in PTEN(+/+) cells via PI3K activity. Collectively, these data in a mouse prostate cell system uncover for the first time a novel and complex relationship between PTEN loss-mediated PI3K/AKT activation and posttranslational regulation of MT1-MMP, which may play a role in PC progression.


Subject(s)
Cell Membrane/metabolism , Matrix Metalloproteinase 14/metabolism , PTEN Phosphohydrolase/metabolism , Protein Processing, Post-Translational , Animals , Cell Line, Tumor , Cell Movement , Enzyme Activation , Enzyme Precursors/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gelatinases/metabolism , Gene Expression Profiling , Genotype , Glycosylation , Humans , Immunoblotting , Male , Matrix Metalloproteinase 14/genetics , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...