Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Cancer Res ; 59(7): 1635-41, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10197640

ABSTRACT

Culturing DOV 13 ovarian carcinoma cells on three-dimensional collagen lattice but not on thin-layer collagen induces processing of promatrix metalloproteinase (MMP)-2 to a M(r) 62,000 form, suggesting that multivalent integrin aggregation may participate in proteinase regulation. To address the role of collagen-binding integrins in this event, we treated DOV 13 cells with soluble beta1 integrin antibodies (clones P4C10 or 21C8) or beta1 integrin antibodies immobilized on latex beads to promote integrin aggregation. Divalent ligation of beta1 integrins with soluble P4C10 antibodies stimulated expression of pro-MMP-2 and its inhibitor, tissue inhibitor of metalloproteinase-2, whereas soluble 21C8 antibodies had no effect. Aggregation of beta1 integrins with immobilized 21C8 or P4C10 antibodies stimulated MMP-dependent pro-MMP-2 activation and accumulation of a M(r) 43,000 form of membrane type 1 MMP (MT1-MMP), a cell surface activator of pro-MMP-2, in cell extracts. beta1 integrin-mediated MMP-2 activation required protein synthesis and tyrosine kinase signaling and was reduced by an inhibitor of gene transcription. Treatment of control cells with concanavalin A stimulated MMP-dependent pro-MMP-2 activation and accumulation of M(r) 55,000 and 43,000 forms of MT1-MMP in cell extracts. Addition of either the MMP inhibitor GM-6001-X or exogenous tissue inhibitor of metalloproteinase-2 to concanavalin A-treated cells resulted in loss of the M(r) 43,000 form of MT1-MMP and accumulation of the M(r) 55,000 form of the enzyme in cell extracts, suggesting that the M(r) 43,000 form is a product of MMP-dependent M(r) 55,000 MT1-MMP proteolysis. Together, these data suggest that beta1 integrin stimulation of pro-MMP-2 activation involves MT1-MMP posttranslational processing and requires multivalent integrin aggregation.


Subject(s)
Gelatinases/biosynthesis , Integrin beta1/physiology , Metalloendopeptidases/biosynthesis , Ovarian Neoplasms/metabolism , Collagen/metabolism , Enzyme Precursors/metabolism , Female , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinases, Membrane-Associated , Molecular Weight , Protease Inhibitors/pharmacology , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tumor Cells, Cultured
2.
Invasion Metastasis ; 16(3): 150-9, 1996.
Article in English | MEDLINE | ID: mdl-9186550

ABSTRACT

Metastatic dissemination of epithelial ovarian carcinoma occurs primarily through exfoliation of cells from the primary tumor, with subsequent implantation, invasion, and growth throughout the organs within the peritoneal cavity. Previous studies have suggested a role for matrix metalloproteinases (MMPs), particularly MMP-2, in ovarian cancer invasion and metastasis. To characterize further the role of MMPs and their inhibitors in ovarian carcinoma, in this study the production and activation of MMPs by short-term primary cultures of human ovarian epithelial carcinoma cells were analyzed. We report that MMP-2 is the predominant gelatinolytic MMP secreted by primary ovarian cancer cells derived from both ovarian tumors and ascites fluid. Furthermore, zymographic analysis demonstrated that MMP-2 is present in conditioned media in both the latent and activated forms, indicating that primary ovarian cancer cells catalyze proMMP-2 activation. Presence of a proMMP-2 activator was confirmed by immunohistochemistry and immunoprecipitation studies which found membrane-type 1 MMP (MT1-MMP) in the membranes of unstimulated cells and levels of both MT1-MMP and tissue inhibitor of metalloproteinases-2 (TIMP-2) were enhanced by culturing cells in the presence of concanavalin A. In addition, interaction of MMP-2 with the ovarian carcinoma cell surface resulted in a 2.5- to 5-fold increase in invasiveness. These data suggest that MT1-MMP-catalyzed activation of proMMP-2 may play a physiologic role in intraperitoneal invasion of ovarian carcinoma cells.


Subject(s)
Carcinoma/enzymology , Gelatinases/biosynthesis , Metalloendopeptidases/biosynthesis , Ovarian Neoplasms/enzymology , Carcinoma/chemistry , Carcinoma/drug therapy , Collagen/metabolism , Concanavalin A/pharmacology , Culture Media, Conditioned , Drug Combinations , Female , Gelatinases/analysis , Humans , Immunohistochemistry , Laminin/metabolism , Matrix Metalloproteinase 2 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/analysis , Neoplasm Invasiveness , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/drug therapy , Proteoglycans/metabolism , Tumor Cells, Cultured
3.
J Biol Chem ; 273(1): 143-9, 1998 Jan 02.
Article in English | MEDLINE | ID: mdl-9417058

ABSTRACT

The initial site of melanoma cell metastasis is frequently the regional lymph nodes, and the appearance of lymph node metastasis correlates with poor prognosis. Lymph node adhesion is mediated by an interaction between the tumor cell integrin alphavbeta3 and lymph node vitronectin. In this study, we explored the relationship between adhesion and proteolysis by examining the direct effect of vitronectin receptor ligation on matrix metalloproteinase-2 (MMP-2) production by B16F1 and B16F10 melanoma cells. We report a dose-dependent increase in secretion of both MMP-2 and tissue inhibitor of metalloproteinases-2 (TIMP-2) in response to vitronectin. Cellular invasiveness was also enhanced by vitronectin, as shown by the increased ability of vitronectin-treated cells to invade a synthetic basement membrane (Matrigel). Both the vitronectin-induced MMP-2 production and vitronectin-enhanced invasion were blocked by the peptide ligand Arg-Gly-Asp-Ser (RGDS). Furthermore, neither plasmin-degraded vitronectin nor the peptide ligand RGDS stimulated MMP-2 secretion or invasiveness, indicating that a multivalent ligand-receptor interaction rather than simple receptor occupancy was required for MMP-2 induction. MMP-2 and MMP-2/TIMP-2 interaction with the plasma membrane of melanoma cells resulted in enhanced catalytic activity against 14C-labeled gelatin, suggesting that membrane association may function in posttranslational regulation of MMP-2 activity. This is supported by data showing increased cellular invasion by cells containing membrane-bound MMP-2. Binding of proMMP-2 and proMMP-2/TIMP-2 to melanoma cells was not inhibited by RGDS, and melanoma cell adhesion to vitronectin was unaffected by pro- or active MMP-2, indicating that MMP-2 did not interact with the murine vitronectin receptor. Together, these data provide evidence for a functional link between adhesion and proteolysis and suggest a potential mechanism whereby adhesion of an invasive cell to the extracellular matrix regulates subsequent invasive behavior.


Subject(s)
Gelatinases/biosynthesis , Melanoma, Experimental/pathology , Metalloendopeptidases/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Vitronectin/metabolism , Animals , Enzyme Induction , Matrix Metalloproteinase 2 , Melanoma, Experimental/enzymology , Melanoma, Experimental/metabolism , Mice , Neoplasm Invasiveness , Tumor Cells, Cultured
4.
Biochem J ; 340 ( Pt 1): 77-84, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10229661

ABSTRACT

Angiostatin, a kringle-containing fragment of plasminogen, is a potent inhibitor of angiogenesis. The mechanism(s) responsible for the anti-angiogenic properties of angiostatin are unknown. We now report that human angiostatin blocks plasmin(ogen)-enhanced in vitro invasion of tissue plasminogen activator (t-PA)-producing endothelial and melanoma cells. Kinetic analyses demonstrated that angiostatin functions as a non-competitive inhibitor of extracellular-matrix (ECM)-enhanced, t-PA-catalysed plasminogen activation, with a Ki of 0.9+/-0.03 microM. This mechanism suggests that t-PA has a binding site for the inhibitor angiostatin, as well as for its substrate plasminogen that, when occupied, prevents ternary complex formation between t-PA, plasminogen and matrix protein. Direct binding experiments confirmed that angiostatin bound to t-PA with an apparent Kd [Kd(app)] of 6.7+/-0.7 nM, but did not bind with high affinity to ECM proteins. Together, these data suggest that angiostatin in the cellular micro-environment can inhibit matrix-enhanced plasminogen activation, resulting in reduced invasive activity, and suggest a biochemical mechanism whereby angiostatin-mediated regulation of plasmin formation could influence cellular migration and invasion.


Subject(s)
Cell Movement/drug effects , Endothelium, Vascular/drug effects , Extracellular Matrix/physiology , Melanoma/pathology , Peptide Fragments/pharmacology , Plasminogen/metabolism , Plasminogen/pharmacology , Angiostatins , Animals , Antibodies/pharmacology , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Extracellular Matrix Proteins/metabolism , Female , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Humans , Kinetics , Melanoma/enzymology , Melanoma/metabolism , Mice , Neovascularization, Pathologic , Peptide Fragments/metabolism , Protein Binding , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/immunology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology , Tumor Cells, Cultured , alpha-2-Antiplasmin/metabolism , alpha-2-Antiplasmin/pharmacology
5.
Cancer ; 80(8): 1457-63, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9338470

ABSTRACT

BACKGROUND: The authors analyzed the secretion of extracellular matrix-degrading proteinases, including urinary-type plasminogen activator (u-PA), matrix metalloproteinase-2 (MMP-2, gelatinase A), and MMP-9 (gelatinase B), by short term primary cultures of epithelial ovarian carcinoma cells derived from primary ovarian tumors, intraperitoneal metastases, or ascites. The presence of these enzymatic activities in samples of ascites was also evaluated. The effect of adhesive substratum on proteinase production was determined. METHODS: A coupled spectrophotometric assay was utilized to evaluate the initial rate of plasminogen activation by u-PA in conditioned medium; this involved monitoring the activity of generated plasmin with a colorimetric substrate. MMP activity was evaluated by gelatin zymography. RESULTS: Ascitic fluids from 18 patients contained u-PA, MMP-2, and MMP-9. However, short term primary cultures of cells derived from primary ovarian tumors (OVET), metastatic lesions (OVEM), or ascites (OVEA) produced very low levels of u-PA. Production of u-PA by OVET and OVEM cells was regulated by adhesive substratum. Conditioned media from OVET, OVEM, and OVEA cells contained high levels of both MMP-2 and MMP-9. MMP-9 levels decreased with increasing passage in culture, whereas MMP-2 activity was maintained. Production of neither MMP-2 nor MMP-9 was regulated by adhesive substratum. CONCLUSIONS: These results demonstrate that primary cultures of epithelial ovarian carcinoma cells derived from three distinct anatomic locations produce MMP-2 and MMP-9, with low level secretion of u-PA. These data suggest that MMPs, particularly MMP-2, may play a significant role in the intraperitoneal invasion of ovarian carcinoma cells.


Subject(s)
Extracellular Matrix/metabolism , Metalloendopeptidases/biosynthesis , Ovarian Neoplasms/enzymology , Plasminogen Activators/biosynthesis , Adult , Aged , Collagenases/biosynthesis , Epithelium/enzymology , Female , Gelatinases/biosynthesis , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Middle Aged , Neoplasm Staging , Tumor Cells, Cultured
6.
Int J Cancer ; 67(5): 695-701, 1996 Sep 04.
Article in English | MEDLINE | ID: mdl-8782661

ABSTRACT

Epithelial ovarian carcinoma, the leading cause of gynecologic cancer death, is characterized by widespread intra-abdominal metastases mediated primarily by surface shedding of tumor cells and peritoneal implantation. Whereas hematogenous metastasis is known to involve cellular adhesion, extracellular matrix proteolysis and cell migration, the role of these processes in the intraperitoneal dissemination of ovarian cancer remains unclear. To analyze further the role of adhesion and proteolysis in ovarian carcinoma dissemination, we have characterized the adhesive profiles of 4 primary cultures of ovarian carcinoma cells and 5 ovarian carcinoma cell lines. Our data demonstrate preferential adhesion of ovarian carcinoma cells to interstitial type I collagen. Analysis of adhesion molecule expression demonstrated the presence of the alpha2 and beta1 integrin subunits by cell surface ELISA, immunoprecipitation and immunohistochemistry. Furthermore, antibodies directed against the alpha2 and beta1 subunits inhibited adhesion of ovarian carcinoma cells to type I collagen by 56% and 95%, respectively. Plasminogen activator and matrix metalloproteinase production by adherent cells was not altered as a consequence of adhesion to individual extracellular matrix proteins; however, adhesion to an extracellular matrix comprised primarily of interstitial collagen increased plasminogen activator activity in 5 of 5 cell lines. Since the ovarian carcinoma micro-environment is rich in type I collagen, our data suggest that preferential adhesion to type I collagen followed by secretion of serine and metalloproteinases may represent a biochemical mechanism by which the intraperitoneal dissemination of ovarian carcinoma is mediated.


Subject(s)
Cell Adhesion , Collagen/metabolism , Integrins/physiology , Ovarian Neoplasms/metabolism , Amino Acid Sequence , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix Proteins/metabolism , Female , Humans , Immunohistochemistry , Immunosorbent Techniques , Metalloendopeptidases/metabolism , Molecular Sequence Data , Muscle, Smooth/physiology , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activators/metabolism , Receptors, Collagen , Serine Endopeptidases/metabolism , Tumor Cells, Cultured
7.
Invasion Metastasis ; 18(1): 15-26, 1998.
Article in English | MEDLINE | ID: mdl-10207247

ABSTRACT

Metastatic dissemination of epithelial ovarian carcinoma is thought to be mediated via tumor cell exfoliation into the peritoneal cavity, followed by adhesion to and invasion through the mesothelium which overlies the contents of the peritoneal cavity. In this study, we have utilized short-term primary cultures to analyze the effect of specific extracellular matrix proteins on properties of human ovarian epithelial carcinoma cells which contribute to the invasive phenotype. Analysis of cell:matrix adhesive profiles indicated that ovarian carcinoma cells adhere preferentially to type I collagen. Immunoprecipitation analyses demonstrated the presence of the collagen-binding alpha2beta1 integrin in biotin-labeled ovarian carcinoma cell membranes, and cellular adhesion was inhibited by blocking antibodies directed against the alpha2 and beta1 integrin subunits. The alpha2beta1-binding peptide Asp-Gly-Glu-Ala (DGEA) was also moderately effective at blocking adhesion to collagen relative to the control peptide Ala-Gly-Glu-Ala (AGEA). Analysis of cell motility on protein-coated colloidal gold coverslips demonstrated that ovarian carcinoma cells migrate preferentially on type I collagen coated surfaces. Type I collagen promoted migration in a concentration-dependent, saturable manner, with maximal migration observed at a collagen-coating concentration of 50 microg/ml. Migration on collagen was inhibited by antibodies directed against the alpha2 and beta1 integrin subunits and by DGEA peptide, providing evidence for the role of the alpha2beta1 integrin in ovarian carcinoma cell motility. Culturing ovarian carcinoma cells on type I collagen gels led to a significant increase in conversion of the matrix metalloproteinase 2 zymogen to the 66-kD form, suggesting that adhesion to collagen also influences matrix-degrading proteinases. These data suggest that alpha2beta1-integrin-mediated interaction of ovarian carcinoma cells with type I collagen, a protein prevalent both in the mesothelial extracellular matrix and in the peritoneal cavity of ovarian carcinoma patients, may function on multiple levels to promote metastatic dissemination of ovarian carcinoma cells.


Subject(s)
Collagen/metabolism , Integrins/metabolism , Neoplasm Metastasis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Cell Adhesion , Cell Movement , Female , Gelatinases/metabolism , Humans , Matrix Metalloproteinase 2 , Metalloendopeptidases/metabolism , Precipitin Tests , Receptors, Collagen , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL