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.
Biochem J ; 441(3): 909-18, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22035541

ABSTRACT

The PDGF (platelet-derived growth factor) family members are potent mitogens for cells of mesenchymal origin and serve as important regulators of cell migration, survival, apoptosis and transformation. Tumour-derived PDGF ligands are thought to function in both autocrine and paracrine manners, activating receptors on tumour and surrounding stromal cells. PDGF-C and -D are secreted as latent dimers, unlike PDGF-A and -B. Cleavage of the CUB domain from the PDGF-C and -D dimers is required for their biological activity. At present, little is known about the proteolytic processing of PDGF-C, the rate-limiting step in the regulation of PDGF-C activity. In the present study we show that the breast carcinoma cell line MCF7, engineered to overexpress PDGF-C, produces proteases capable of cleaving PDGF-C to its active form. Increased PDGF-C expression enhances cell proliferation, anchorage-independent cell growth and tumour cell motility by autocrine signalling. In addition, MCF7-produced PDGF-C induces fibroblast cell migration in a paracrine manner. Interestingly, PDGF-C enhances tumour cell invasion in the presence of fibroblasts, suggesting a role for tumour-derived PDGF-C in tumour-stromal interactions. In the present study, we identify tPA (tissue plasminogen activator) and matriptase as major proteases for processing of PDGF-C in MCF7 cells. In in vitro studies, we also show that uPA (urokinase-type plasminogen activator) is able to process PDGF-C. Furthermore, by site-directed mutagenesis, we identify the cleavage site for these proteases in PDGF-C. Lastly, we provide evidence suggesting a two-step proteolytic processing of PDGF-C involving creation of a hemidimer, followed by GFD-D (growth factor domain dimer) generation.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Serine Proteases/physiology , Animals , Autocrine Communication/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Lymphokines/antagonists & inhibitors , Lymphokines/genetics , Lymphokines/physiology , Mice , NIH 3T3 Cells , Paracrine Communication/genetics , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/physiology , Protein Multimerization/genetics , Protein Multimerization/physiology , Protein Processing, Post-Translational , Serine Proteases/genetics , Serine Proteases/metabolism , Transfection
2.
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
3.
Traffic ; 9(2): 215-29, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18034774

ABSTRACT

Human adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 1 (APPL1) and adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 2 (APPL2) are homologous effectors of the small guanosine triphosphatase RAB5 that interact with a diverse set of receptors and signaling proteins and are proposed to function in endosome-mediated signaling. Herein, we investigated the membrane-targeting properties of the APPL1 and APPL2 Bin/Amphiphysin/Rvs (BAR), pleckstrin homology (PH) and phosphotyrosine binding (PTB) domains. Coimmunoprecipitation and yeast two-hybrid studies demonstrated that full-length APPL proteins formed homooligomers and heterooligomers and that the APPL minimal BAR domains were necessary and sufficient for mediating APPL-APPL interactions. When fused to a fluorescent protein and overexpressed, all three domains (minimal BAR, PH and PTB) were targeted to cell membranes. Furthermore, full-length APPL proteins bound to phosphoinositides, and the APPL isolated PH or PTB domains were sufficient for in vitro phosphoinositide binding. Live cell imaging showed that full-length APPL-yellow fluorescent protein (YFP) fusion proteins associated with cytosolic membrane structures that underwent movement, fusion and fission events. Overexpression of full-length APPL-YFP fusion proteins was sufficient to recruit endogenous RAB5 to enlarged APPL-associated membrane structures, although APPL1 was not necessary for RAB5 membrane targeting. Taken together, our findings suggest a role for APPL proteins as dynamic scaffolds that modulate RAB5-associated signaling endosomal membranes by their ability to undergo domain-mediated oligomerization, membrane targeting and phosphoinositide binding.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Membrane/metabolism , Phosphatidylinositols/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Cytoplasmic Vesicles/metabolism , Golgi Apparatus/metabolism , Humans , Immunoprecipitation , Intracellular Membranes/metabolism , Mice , Protein Binding , Protein Transport/physiology , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Two-Hybrid System Techniques , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
4.
Mol Cell Biol ; 25(14): 6279-88, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988036

ABSTRACT

Platelet-derived growth factor (PDGF) protein family members are potent mitogens and chemoattractants for mesenchymal cells. The classic PDGF ligands A and B are single-domain protein chains which are secreted as active dimers capable of activating their cognate PDGF receptors (PDGFRs). In contrast to PDGFs A and B, PDGF D contains an N-terminal complement subcomponent C1r/C1s, Uegf, and Bmp1 (CUB) domain and a C-terminal PDGF domain. PDGF D must undergo extracellular proteolytic processing, separating the CUB domain from the PDGF domain, before the PDGF domain can stimulate beta-PDGFR-mediated cell signal transduction. Here, we report that prostate carcinoma cells LNCaP and PC3 autoactivate latent full-length PDGF D into its active form under serum-independent conditions and that this autoactivation is inhibited by PAI-1, a urokinase plasminogen activator (uPA)/tissue plasminogen activator (tPA) inhibitor. Interestingly, uPA, but not the closely related protease tPA, is capable of processing recombinant latent PDGF DD into the active form. We identify the uPA cleavage site between the CUB and PDGF domains of the full-length PDGF D by mutational analysis and show that PDGF D and uPA colocalize in human prostate carcinoma. This evidence provides a direct link between uPA- and PDGF D-mediated cell signaling, which may contribute to the progression of prostate cancer.


Subject(s)
Carcinoma/enzymology , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/enzymology , Urokinase-Type Plasminogen Activator/metabolism , Amino Acid Sequence , Aprotinin/pharmacology , Carcinoma/chemistry , Cell Line, Tumor , DNA Mutational Analysis , Humans , Lymphokines/analysis , Lymphokines/genetics , Male , Molecular Sequence Data , Plasminogen Activator Inhibitor 1/pharmacology , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/genetics , Prostatic Neoplasms/chemistry , Urokinase-Type Plasminogen Activator/analysis , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
5.
Cancer Res ; 64(5): 1722-9, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14996732

ABSTRACT

The platelet-derived growth factor (PDGF) proteins are potent stimulators of cell proliferation/transformation and play a major role in cell-cell communication. For over two decades, PDGFs were thought to exist as three dimeric polypeptides (the homodimers AA and BB and the heterodimer AB). Recently, however, the PDGF C and D chains were discovered in a BLAST search of the expressed sequence tag databases. The PDGF CC and DD dimers have a unique two-domain structure with an NH(2)-terminal CUB (compliment subcomponents C1r/C1s, Uegf, and Bmp1) domain and a COOH-terminal PDGF/vascular endothelial growth factor domain. Whereas secreted PDGF AA, BB, and AB readily activate their cell surface receptors, it was suggested that extracellular proteolytic removal of the CUB domain is required for the PDGF/vascular endothelial growth factor domain of PDGF CC and DD to activate PDGF receptors. In the present study, we examined the processing of latent PDGF D into its active form and the effects of PDGF D expression on prostate cancer progression. We show that LNCaP cells auto-activate latent PDGF DD into the active PDGF domain, which can induce phosphorylation of the beta-PDGF receptor and stimulates LNCaP cell proliferation in an autocrine manner. Additionally, LNCaP-PDGF D-conditioned medium induces migration of the prostate fibroblast cell line 1532-FTX, indicating LNCaP-processed PDGF DD is active in a paracrine manner as well. In a severe combined immunodeficient mouse model, PDGF DD expression accelerates early onset of prostate tumor growth and drastically enhances prostate carcinoma cell interaction with surrounding stromal cells. These demonstrate a potential oncogenic activity of PDGF DD in the development and/or progression of prostate cancer.


Subject(s)
Lymphokines , Platelet-Derived Growth Factor/physiology , Prostatic Neoplasms/pathology , Cell Communication , Cell Division , Cell Movement , Disease Progression , Humans , Male , Prostatic Neoplasms/etiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Stromal Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...