Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Microvasc Res ; 75(2): 169-78, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17976661

ABSTRACT

The matrix-inserted surface transplantation model is an in vivo assay used to analyse the kinetics of tumor-vessel interactions during different stages of skin carcinoma progression. This system allows the study of host-tumor interface, i.e. penetration of tumor cells into normal host tissue as well as infiltration of normal host cells into the tumor. In the present study, image analysis algorithms for processing and quantifying the extent of such migratory and tissue remodeling events are presented. The proposed method is non-parametric and its originality lies in its particularity to take into account the specific geometry of tumor-host interface. This methodology is validated by evaluating the contribution of matrix metalloproteases (MMPs) in skin carcinoma invasion and vascularization through pharmacological and genetic approaches.


Subject(s)
Neovascularization, Pathologic/pathology , Signal Processing, Computer-Assisted , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Algorithms , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Cell Movement , Dipeptides/pharmacology , Dipeptides/therapeutic use , Kinetics , Matrix Metalloproteinases, Secreted/antagonists & inhibitors , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Statistical , Neoplasm Invasiveness , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/prevention & control , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Reproducibility of Results , Skin Neoplasms/enzymology , Skin Neoplasms/prevention & control , Stromal Cells/pathology
2.
J Thromb Haemost ; 5(9): 1936-44, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17723133

ABSTRACT

BACKGROUND: Degradation of extracellular matrix proteins, such as fibrin, is pivotal to tumor invasion. Inhibition of the interaction between urokinase plasminogen activator (u-PA) and its receptor (u-PAR), and hence pro-u-PA activation, is an attractive approach to anti-invasive cancer therapy. A number of inhibitors exist for the human system, but because of species specificity none of these are efficient in mice. We have recently generated an inhibitory monoclonal antibody (mAb) against mouse u-PAR (mR1) by immunization of u-PAR-deficient mice. OBJECTIVES: To evaluate the effect of mR1 in vivo in a physiological setting sensitive to deregulated fibrinolysis, we have administered mR1 systemically and quantitated the effect on liver fibrin accumulation. METHODS: Wild-type and tissue-type plasminogen activator (t-PA) deficient mice were administered with mR1, or control antibody, during 6 weeks. Thereafter, the livers were retrieved and the amount of liver fibrin measured by unbiased morphometrical analysis of immunofluorescence signal. RESULTS: Systemic administration of mR1 caused significantly increased fibrin signal in anti-u-PAR treated t-PA-deficient mice compared to mock-treated, which mimics the phenotype of u-PAR;t-PA double-deficient mice. Fibrin and fibronectin accumulated within the sinusoidal space and was infiltrated by inflammatory cells. Analysis of small and rare hepatic fibrin plaques observed in t-PA-deficient mice showed infiltrating macrophages that, contrary to surrounding Kuppfer cells, expressed u-PAR. CONCLUSION: We show that u-PAR-expressing macrophages are involved in cell-mediated fibrinolysis of liver fibrin deposits, and that the antimouse-u-PAR mAb is effective in vivo and thus suited for studies of the effect of targeting the u-PA/u-PAR interaction in mouse cancer models.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Fibrin/metabolism , Liver/drug effects , Receptors, Cell Surface/immunology , Tissue Plasminogen Activator/genetics , Animals , Antibodies, Monoclonal/pharmacology , Fluorescent Antibody Technique , Liver/metabolism , Mice , Mice, Inbred C57BL , Receptors, Urokinase Plasminogen Activator
3.
Scand J Immunol ; 58(3): 298-305, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12950675

ABSTRACT

The urokinase receptor (uPAR) is a glycolipid-anchored cell surface glycoprotein that plays a central role in extracellular proteolysis during tissue remodeling processes including cancer invasion. Furthermore, uPAR is found on the surface of both dendritic cells (DCs) and T cells, and has been proposed to play a role in DC-induced T-cell activation and, therefore, in the induction of an immune response. In order to investigate the possibility of using DNA immunization for the generation of poly- and monoclonal antibodies to uPAR, we injected wild-type mice and mice deficient in uPAR (uPAR knockouts) intramuscularly with plasmid DNA encoding a carboxy-terminal truncated soluble form of the human uPAR. Multiple injections of 100 micro g of DNA resulted in a strong and specific antibody response in all mice irrespective of genotype. Antisera with a maximum titre of 32,000 were obtained, comparable with that obtained after immunization with recombinant uPAR. The subclass distribution of uPAR-specific antibodies in the sera demonstrated the induction of a mixed TH1/TH2 response, irrespective of the genotype of the mice. Our results demonstrate the possibility of generating high titre antibodies to uPAR by DNA immunization of wild-type as well as uPAR knockout mice, and that cell surface uPAR is not indispensable for the generation of a humoral immune response.


Subject(s)
Antibody Formation/immunology , Receptors, Cell Surface/immunology , Vaccines, DNA/immunology , Animals , Antibodies/blood , Antibody Formation/genetics , Blotting, Western , COS Cells , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors/genetics , Humans , Immunization , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Immunoglobulin G/immunology , Mice , Mice, Knockout , Plasmids/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Transfection , Vaccines, DNA/genetics
4.
Placenta ; 24(6): 677-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12828926

ABSTRACT

Clearance of fibrin deposits within the human placenta is an ongoing process during normal placental development. Plasminogen is a circulating fibrinolytic protease zymogen activated in situ by plasminogen activators. We have previously reported that the receptor for urokinase plasminogen activator (uPAR) is expressed by cells either covering or enmeshed within the perivillous fibrinoid deposits. Whereas these cells seemed likely to be trophoblasts, a definitive identification was lacking, and this question is central to the understanding of the cellular mechanisms directing fibrinolysis in the placenta. In this study we have performed immunohistochemical co-localization studies and found that the uPAR-positive cells covering fibrinoid deposits are immunoreactive for CD31 and vWF, indicating that they are actually endothelial cells. In addition, we found that perivillous fibrinoid deposits not covered with uPAR-positive endothelial cells were covered with platelets identified by integrin alpha(IIb)beta(3)-immunoreactivity. Also surprisingly, the uPAR-positive cells enmeshed within fibrinoid deposits express a cell specific marker indicating that they are macrophages. Both uPAR-positive cell populations also express uPA immunoreactivity. Taken together, the data suggest that both fibrinoid-covering endothelial cells and fibrinoid-enmeshed macrophages can participate in the clearance process of perivillous fibrinoid deposits formed in the human placenta.


Subject(s)
Endothelium/metabolism , Fibrin/metabolism , Macrophages/metabolism , Placenta/metabolism , Receptors, Cell Surface/metabolism , Adult , Biomarkers/analysis , Blood Platelets/metabolism , Endothelium/cytology , Female , Humans , Macrophages/cytology , Placenta/cytology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Pregnancy , Receptors, Urokinase Plasminogen Activator , Up-Regulation , von Willebrand Factor/analysis
5.
Cancer Res ; 61(19): 7091-100, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585740

ABSTRACT

Collagenase-3 (matrix metalloproteinase 13; MMP-13), a protease originally identified in breast carcinoma, is characterized by a potent degrading activity against a wide spectrum of extracellular matrix proteins. The aims of this study were to localize and identify the MMP-13-expressing cells in invasive human breast carcinoma and to evaluate the role of MMP-13 in transition to invasive lesions by studying ductal carcinoma in situ (DCIS). We found expression of MMP-13 in stromal fibroblast-like cells in all 21 invasive ductal carcinomas studied and in 4 of 9 invasive lobular carcinomas. In most carcinomas, expression of MMP-13 was limited to small stromal foci in the tumor area. Combined in situ hybridization and immunohistochemistry showed coexpression of alpha-smooth muscle actin immunoreactivity and MMP-13 mRNA in myofibroblasts. In contrast, cytokeratin-positive cancer cells, alpha-smooth muscle actin-positive vascular smooth muscle cells, CD68-positive macrophages, and CD31-positive endothelial cells were all MMP-13 mRNA negative. In situ hybridization for MMP-13 in 17 DCIS lesions revealed expression in 10 cases. Immunohistochemical analysis of all DCIS cases identified microinvasion in 8 of the 17 lesions. Seven of the eight lesions with microinvasion were MMP-13 positive. Further analysis showed that MMP-13 expression was often associated with the microinvasive events. This particular expression pattern was unique for MMP-13 among other MMPs analyzed, including MMP-2, -11, and -14. We conclude that MMP-13 is primarily expressed by myofibroblasts in human breast carcinoma and that expression in DCIS lesions often is associated with microinvasive events. On the basis of these data, we propose that MMP-13 may play an essential role during transition of DCIS lesions to invasive ductal carcinomas.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma in Situ/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Lobular/enzymology , Collagenases/biosynthesis , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Disease Progression , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Matrix Metalloproteinase 13 , Neoplasm Invasiveness
6.
Lab Invest ; 81(10): 1403-14, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11598153

ABSTRACT

The urokinase plasminogen activator receptor-associated protein/Endo180 (uPARAP/Endo180) is a newly discovered member of the macrophage mannose receptor family that was reported to interact with ligand-bound urokinase plasminogen activator receptor (uPAR), matrix metalloprotease-13 (MMP-13), and collagen V on the cell surface. We have determined the sites of expression of this novel receptor during murine postimplantation development. uPARAP/Endo180 was expressed in all tissues undergoing primary ossification, including the developing bones of the viscerocranium and calvarium that ossify intramembranously, and developing long bones undergoing endochondral ossification. uPARAP/Endo180 mRNA was expressed by both immature osteoblasts and by mature osteocalcin-producing osteoblasts-osteocytes, and was coexpressed with MMP-13. Interestingly, osteoblasts also expressed uPAR. Besides bone-forming tissues, uPARAP/Endo180 expression was detected only in a mesenchymal condensation of the midbrain and in the developing lungs. The data suggest a function of this novel protease receptor in bone development, possibly mediated through its interactions with uPAR, MMP-13, or collagen V.


Subject(s)
Bone and Bones/physiology , Collagenases/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Mitogen/biosynthesis , Animals , Bone and Bones/embryology , Embryonic and Fetal Development , Female , Immunohistochemistry , Matrix Metalloproteinase 13 , Mice , Osteogenesis/physiology , Pregnancy , Receptors, Urokinase Plasminogen Activator
7.
Am J Pathol ; 158(6): 1997-2003, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395377

ABSTRACT

We have developed a computer-assisted stereological method based on unbiased principles for estimating metastasis volumes in mouse lungs. We evaluated this method using the transplantable Lewis lung carcinoma. Twenty-one days after subcutaneous inoculation of 10(6) Lewis lung cells into C57BL/6J mice, the mice had primary tumors with an average volume of 2300 mm(3). After perfusion fixation, the lungs were removed, embedded in OCT compound, snap-frozen, and processed for stereology. The metastasis volumes were estimated by application of the Cavalieri principle after evaluation of single sections from several evenly distributed tissue levels. The metastasis volume in a group of nine mice varied between 0.01 and 14.4 mm(3), with an average of 6.1 mm(3). The coefficient of variation was 0.9. The coefficient of error of the volume estimation was determined in five cases and varied from 0.08 to 0.23. Thus, the variation on the metastasis volumes that is achieved by this method contributes very little, 2.5%, to the total variance within the group of mice. In conclusion, we have developed an efficient and unbiased method to determine the metastasis burden in mouse lungs.


Subject(s)
Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/secondary , Image Processing, Computer-Assisted/methods , Neoplasm Metastasis/pathology , Animals , Cell Division , Female , Kinetics , Lung/pathology , Mice , Mice, Inbred C57BL , Organ Size , Tissue Preservation
8.
J Exp Med ; 193(7): 793-802, 2001 Apr 02.
Article in English | MEDLINE | ID: mdl-11283152

ABSTRACT

Matrix metalloproteinase (MMP)9/gelatinase B is increased in various nephropathies. To investigate its role, we used a genetic approach. Adult MMP9-deficient (MMP9(-/)-) mice showed normal renal histology and function at 3 mo. We investigated the susceptibility of 3-mo-old mice to the accelerated model of anti-glomerular basement membrane nephritis, in which fibrin is an important mediator of glomerular injury and renal impairment. Unexpectedly, nephritis was more severe in MMP9(-/)- than in control mice, as attested by levels of serum creatinine and albuminuria, and the extent of crescents and fibrin deposits. Circulating or deposited immunoglobulin G, interleukin (IL)-1beta, or IL-10 were the same in MMP9(-/-) and MMP9(+/+) mice. However, we found that fibrin is a critical substrate for MMP9, and in its absence fibrin accumulated in the glomeruli. These data indicate that MMP9 is required for a novel protective effect on the development of fibrin-induced glomerular lesions.


Subject(s)
Anti-Glomerular Basement Membrane Disease/etiology , Fibrin/metabolism , Kidney Glomerulus/pathology , Matrix Metalloproteinase 9/metabolism , Animals , Anti-Glomerular Basement Membrane Disease/pathology , Basement Membrane/immunology , Kidney Function Tests , Matrix Metalloproteinase 9/genetics , Mice , Mice, Mutant Strains , Proteinuria
9.
J Cell Biol ; 152(4): 777-84, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11266468

ABSTRACT

The plasminogen (Plg)/plasminogen activator (PA) system plays a key role in cancer progression, presumably via mediating extracellular matrix degradation and tumor cell migration. Consequently, urokinase-type PA (uPA)/plasmin antagonists are currently being developed for suppression of tumor growth and angiogenesis. Paradoxically, however, high levels of PA inhibitor 1 (PAI-1) are predictive of a poor prognosis for survival of patients with cancer. We demonstrated previously that PAI-1 promoted tumor angiogenesis, but by an unresolved mechanism. We anticipated that PAI-1 facilitated endothelial cell migration via its known interaction with vitronectin (VN) and integrins. However, using adenoviral gene transfer of PAI-1 mutants, we observed that PAI-1 promoted tumor angiogenesis, not by interacting with VN, but rather by inhibiting proteolytic activity, suggesting that excessive plasmin proteolysis prevents assembly of tumor vessels. Single deficiency of uPA, tissue-type PA (tPA), uPA receptor, or VN, as well as combined deficiencies of uPA and tPA did not impair tumor angiogenesis, whereas lack of Plg reduced it. Overall, these data indicate that plasmin proteolysis, even though essential, must be tightly controlled during tumor angiogenesis, probably to allow vessel stabilization and maturation. These data provide insights into the clinical paradox whereby PAI-1 promotes tumor progression and warrant against the uncontrolled use of uPA/plasmin antagonists as tumor angiogenesis inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endopeptidases/metabolism , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Plasminogen Activator Inhibitor 1/pharmacology , Vitronectin/metabolism , Animals , Endothelium, Vascular/drug effects , Fibrinolysin/metabolism , Keratinocytes/pathology , Mice , Mice, Mutant Strains , Muscle Neoplasms/blood supply , Neoplasm Invasiveness , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/etiology , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Vitronectin/genetics
10.
J Invest Dermatol ; 116(3): 353-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231307

ABSTRACT

In this study we have used in situ hybridization with radiolabeled antisense RNA probes to examine the expression of mRNA for urokinase-type plasminogen activator and its receptor in histologic samples of squamous cell (n = 7) and basal cell (n = 7) carcinomas of the skin. Messenger RNA for both urokinase-type plasminogen activator and its receptor were expressed in all of the squamous cell carcinomas, but could not be detected in the basal cell carcinomas. In all of the seven squamous cell carcinomas a signal for urokinase-type plasminogen activator receptor mRNA was detected focally in well-differentiated cancer cells surrounding keratinized pearls, and in four specimens urokinase-type plasminogen activator receptor mRNA was in addition expressed by cancer cells at the edge of invasively growing strands of tumor. Urokinase-type plasminogen activator mRNA expression was found in virtually all the cancer cells of the squamous cell carcinomas, and importantly we found, by hybridizations for urokinase-type plasminogen activator and its receptor mRNA on adjacent sections of squamous cell carcinomas, that it was exactly the invading cancer cells that simultaneously expressed both these components required for plasmin-mediated proteolysis at the cell surface. We have previously shown that both urokinase-type plasminogen activator and its receptor mRNA are expressed by the leading-edge keratinocytes in regenerating epidermis during mouse skin wound healing, and that wound healing is impaired in mice made deficient in plasminogen by targeted gene disruption. We propose that there are similarities between the mechanisms of generation and regulation of extracellular proteolysis during skin re-epithelialization and squamous cell carcinoma invasion. The ability of the squamous carcinoma cells to mimic the "invasive" phenotype of re-epithelializing keratinocytes may be one of the factors that make squamous cell carcinomas more aggressive tumors than basal cell carcinomas.


Subject(s)
Carcinoma, Squamous Cell/metabolism , RNA, Messenger/metabolism , Skin Neoplasms/metabolism , Urokinase-Type Plasminogen Activator/genetics , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Humans , Neoplasm Invasiveness , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Skin Neoplasms/pathology
11.
Nat Cell Biol ; 3(3): 267-75, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231576

ABSTRACT

Here we show that plasma kallikrein (PKal) mediates a plasminogen (Plg) cascade in adipocyte differentiation. Ecotin, an inhibitor of serine proteases, inhibits cell-shape change, adipocyte-specific gene expression, and lipid accumulation during adipogenesis in culture. Deficiency of Plg, but not of urokinase or tissue-type plasminogen activator, suppresses adipogenesis during differentiation of 3T3-L1 cells and mammary-gland involution. PKal, which is inhibited by ecotin, is required for adipose conversion, Plg activation and 3T3-L1 differentiation. Human plasma lacking PKal does not support differentiation of 3T3-L1 cells. PKal is therefore a physiological regulator that acts in the Plg cascade during adipogenesis. We propose that the Plg cascade fosters adipocyte differentiation by degradation of the fibronectin-rich preadipocyte stromal matrix.


Subject(s)
Adipocytes/cytology , Cell Differentiation/physiology , Coagulants/metabolism , Escherichia coli Proteins , Periplasmic Proteins , Plasma Kallikrein/metabolism , Plasminogen/metabolism , Adipocytes/physiology , Animals , Azo Compounds/metabolism , Bacterial Proteins/pharmacology , Blotting, Western , Cells, Cultured , Coloring Agents/metabolism , Culture Media, Serum-Free , Female , Fibrinolysin/metabolism , Fibronectins/metabolism , Humans , Immunohistochemistry , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/cytology , Mice , Serine Proteinase Inhibitors/pharmacology
12.
J Histochem Cytochem ; 49(2): 237-46, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11156692

ABSTRACT

uPAR is a cellular receptor for urokinase plasminogen activator, an enzyme involved in extracellular matrix degradation during processes involving tissue remodeling. We have expressed a recombinant soluble form of murine uPAR and raised rabbit polyclonal antibodies to study the expression of uPAR by immunohistochemistry. The immunohistochemical localization of uPAR was determined in normal mouse organs and in tumors formed by the highly metastatic Lewis lung carcinoma. uPAR immunoreactivity was found in the lungs, kidneys, and spleen, and in endothelial cells in the uterus, urinary bladder, thymus, heart, liver, and testis. No uPAR immunoreactivity was detected in muscle. In general, strong uPAR immunoreactivity was observed in organs undergoing extensive tissue remodeling, as exemplified by trophoblast cells in placenta, and in migrating, but not resting, keratinocytes at the edge of incisional wounds. Staining was not detected in any tissue sections derived from uPAR-deficient mice, thus confirming the specificity of the immunohistochemical staining of uPAR in normal mouse tissues. In Lewis lung carcinoma, uPAR immunoreactivity was found in the tumor cells of the primary tumor and in lung metastases. (J Histochem Cytochem 49:237-246, 2001)


Subject(s)
Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antibodies , Blotting, Western , CHO Cells , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cricetinae , Cross-Linking Reagents , Female , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Organ Specificity , Rabbits , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Transfection
13.
J Cell Biol ; 151(4): 879-89, 2000 Nov 13.
Article in English | MEDLINE | ID: mdl-11076971

ABSTRACT

Bone development requires the recruitment of osteoclast precursors from surrounding mesenchyme, thereby allowing the key events of bone growth such as marrow cavity formation, capillary invasion, and matrix remodeling. We demonstrate that mice deficient in gelatinase B/matrix metalloproteinase (MMP)-9 exhibit a delay in osteoclast recruitment. Histological analysis and specialized invasion and bone resorption models show that MMP-9 is specifically required for the invasion of osteoclasts and endothelial cells into the discontinuously mineralized hypertrophic cartilage that fills the core of the diaphysis. However, MMPs other than MMP-9 are required for the passage of the cells through unmineralized type I collagen of the nascent bone collar, and play a role in resorption of mineralized matrix. MMP-9 stimulates the solubilization of unmineralized cartilage by MMP-13, a collagenase highly expressed in hypertrophic cartilage before osteoclast invasion. Hypertrophic cartilage also expresses vascular endothelial growth factor (VEGF), which binds to extracellular matrix and is made bioavailable by MMP-9 (Bergers, G., R. Brekken, G. McMahon, T.H. Vu, T. Itoh, K. Tamaki, K. Tanzawa, P. Thorpe, S. Itohara, Z. Werb, and D. Hanahan. 2000. Nat. Cell Biol. 2:737-744). We show that VEGF is a chemoattractant for osteoclasts. Moreover, invasion of osteoclasts into the hypertrophic cartilage requires VEGF because it is inhibited by blocking VEGF function. These observations identify specific actions of MMP-9 and VEGF that are critical for early bone development.


Subject(s)
Bone Development/physiology , Endothelial Growth Factors/physiology , Lymphokines/physiology , Matrix Metalloproteinase 9/metabolism , Osteoclasts/physiology , Animals , Bone Development/drug effects , Bone Resorption , Chemotaxis , Crosses, Genetic , Heterozygote , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/drug effects , Protease Inhibitors/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Development ; 127(20): 4481-92, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003846

ABSTRACT

Urokinase-type plasminogen activator expression is induced in the mouse mammary gland during development and post-lactational involution. We now show that primiparous plasminogen-deficient (Plg(-/-)) mice have seriously compromised mammary gland development and involution. All mammary glands were underdeveloped and one-quarter of the mice failed to lactate. Although the glands from lactating Plg(-/-) mice were initially smaller, they failed to involute after weaning, and in most cases they failed to support a second litter. Alveolar regression was markedly reduced and a fibrotic stroma accumulated in Plg(-/-) mice. Nevertheless, urokinase and matrix metalloproteinases (MMPs) were upregulated normally in involuting glands of Plg(-/-) mice, and fibrin did not accumulate in the glands. Heterozygous Plg(+/-) mice exhibited haploinsufficiency, with a definite, but less severe mammary phenotype. These data demonstrate a critical, dose-dependent requirement for Plg in lactational differentiation and mammary gland remodeling during involution.


Subject(s)
Lactation/physiology , Mammary Glands, Animal/physiology , Plasminogen/deficiency , Animals , Cell Differentiation , Extracellular Matrix/metabolism , Female , Fertility/physiology , Fibrin/metabolism , Mammary Glands, Animal/cytology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Mutant Strains , Plasminogen/genetics , Urokinase-Type Plasminogen Activator/metabolism
15.
Placenta ; 21(2-3): 170-6, 2000.
Article in English | MEDLINE | ID: mdl-10736239

ABSTRACT

An intimately regulated cell surface activation of matrix metalloproteinases (MMPs) is believed to be of critical importance for the control of trophoblast invasion. A histological investigation of the expression and localization of three different MMPs, the membrane-type matrix metalloproteinases 1 and 2 (MT1-MMP, MT2-MMP) and matrix metalloproteinase 2 (MMP-2/gelatinase A) was performed by in situ hybridization on consecutive sections from human placentae of first trimester pregnancies. Cytokeratin immunostaining identified trophoblast cells. Both normal and tubal implantation sites were studied. We observed a high degree of coexpression of MT2-MMP, MT1-MMP and MMP-2 mRNAs in single extravillous cytotrophoblasts that had invaded the endometrium and tubal wall. Furthermore, mRNAs for all three genes were also seen in cytotrophoblasts of cell islands. In contrast to this coexpression pattern, MT2-MMP expression was absent from cell columns and decidual cells, in which signals for MT1-MMP and MMP-2 mRNAs were seen. The present data on the cellular expression of MT2-MMP mRNA in placenta extend our knowledge of the proteolytic events that take place during early pregnancy. The data suggest that MT2-MMP, capable of activating MMP-2 in vitro, is involved in the invasion of extravillous cytotrophoblast, possibly related to the physiological activation of MMP-2.


Subject(s)
Metalloendopeptidases/genetics , Placenta/enzymology , RNA, Messenger/genetics , Enzyme Activation , Female , Gene Expression , Humans , In Situ Hybridization , Matrix Metalloproteinase 15 , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/metabolism , Pregnancy , Pregnancy Trimester, First , Pregnancy, Tubal/enzymology , Pregnancy, Tubal/genetics , RNA, Messenger/metabolism , Trophoblasts/enzymology
16.
Biochemistry ; 39(3): 508-15, 2000 Jan 25.
Article in English | MEDLINE | ID: mdl-10642175

ABSTRACT

The plasminogen activation (PA) system is involved in the degradation of fibrin and various extracellular matrix proteins, taking part in a number of physiological and pathological tissue remodeling processes including cancer invasion. This system is organized as a classical proteolytic cascade, and as for other cascade systems, understanding the physiological initiation mechanism is of central importance. The attempts to identify initiation routes for activation of the proform of the key enzyme urokinase-type plasminogen activator (pro-uPA) in vivo have been hampered by the strong activator potency of the plasmin, that is generated during the progress of the cascade. Using gene-targeted mice deficient in plasminogen (Plg -/- mice) [Bugge, T. H., Flick, M. J., Daugherty, C. C., and Degen, J. L. (1995) Genes Dev. 9, 794-807], we have now demonstrated and identified a component capable of initiating the cascade by activating pro-uPA. The urine from Plg -/- mice contained active two-chain uPA as well as a proteinase capable of activating exogenously added pro-uPA. The active component was purified and identified by mass spectrometry-based peptide mapping as mouse glandular kallikrein mGK-6 (true tissue kallikrein). The pro-uPA converting activity of the mGK-6 enzyme, as well as its ability to cleave a synthetic substrate for glandular kallikrein, was inhibited by the serine proteinase inhibitor leupeptin but not by other serine proteinase inhibitors such as aprotinin, antithrombin III, or alpha(1)-antitrypsin. We suggest that mouse glandular kallikrein mGK-6 is an activator of pro-uPA in the mouse urinary tract in vivo. Since this kallikrein is expressed in a number of tissues and also occurs in plasma, it can also be considered a candidate for a physiological pro-uPA activator in other locations.


Subject(s)
Plasminogen/metabolism , Tissue Kallikreins/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Amino Acid Sequence , Animals , Enzyme Activation , Enzyme Precursors/metabolism , Fibrinolysin/metabolism , Humans , Macromolecular Substances , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Molecular Sequence Data , Peptide Fragments/chemistry , Plasminogen/deficiency , Plasminogen/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thrombin/metabolism , Tissue Kallikreins/isolation & purification , Tissue Kallikreins/urine , Urokinase-Type Plasminogen Activator/chemistry
17.
EMBO J ; 18(17): 4645-56, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10469644

ABSTRACT

Retarded wound healing was found in mice deficient in the serine protease precursor plasminogen, as well as in wild-type mice treated with the metalloprotease inhibitor galardin, but in both cases wound closure was ultimately completed in all mice within 60 days. The expression of several matrix metalloproteases in keratinocytes migrating to cover the wound was strongly enhanced by galardin treatment. However, when plasminogen-deficient mice were treated with galardin, healing was completely arrested and wound closure was not seen during an observation period of 100 days, demonstrating that protease activity is essential for skin wound healing. The requirement for both plasminogen deficiency and metalloprotease inhibition for complete inhibition of the healing process indicates that there is a functional overlap between the two classes of matrix-degrading proteases, probably in the dissection of the fibrin-rich provisional matrix by migrating keratinocytes. Each class alone is capable of maintaining sufficient keratinocyte migration to regenerate the epidermal surface, although this function would normally be performed by both classes acting in parallel. Since there are strong similarities between the proteolytic mechanisms in wound healing and cancer invasion, these results predict that complete arrest of this latter process in therapeutic settings will require the use of inhibitors of both classes of proteases.


Subject(s)
Metalloendopeptidases/metabolism , Wound Healing/physiology , Animals , Cell Movement/drug effects , Dipeptides/pharmacology , Gene Expression Regulation, Enzymologic , Immunohistochemistry , Keratinocytes/drug effects , Metalloendopeptidases/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Plasminogen/genetics , Plasminogen/physiology , Protease Inhibitors/pharmacology , Skin/anatomy & histology , Skin/enzymology , Time Factors , Wound Healing/drug effects
18.
APMIS ; 107(1): 120-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10190288

ABSTRACT

Proteolytic degradation of the extracellular matrix plays a crucial role in both cancer invasion and non-neoplastic tissue remodeling processes. In human cancers the components of matrix degrading protease systems (uPA, uPAR, PAI-1 and MMPs) can be expressed by either the non-neoplastic stromal cells, the cancer cells or both. Studies of the prognostic impact of these components in human cancer and the effect of targeted gene inactivation on cancer metastasis in mice support the assumption that proteases promote cancer progression, independent of whether they are expressed by cancer cells or stromal cells. The pattern of expression of components of protease systems is usually very similar in different cases of the same type of cancer, while it varies between different types of cancer. There are intriguing similarities between the cellular expression pattern of components of protease systems seen in cancer invasion and in certain types of non-neoplastic tissue remodeling. We propose that cancer invasion can be viewed as tissue remodeling gone out of control. The stromal cell involvement in cancer invasion represents a new paradigm with important implications for cancer pathophysiology and cancer therapy.


Subject(s)
Endopeptidases/physiology , Extracellular Matrix/metabolism , Neoplasm Invasiveness , Animals , Humans , Mice , Plasminogen Activator Inhibitor 1/physiology , Receptors, Cell Surface/physiology , Receptors, Urokinase Plasminogen Activator , Stromal Cells/physiology , Wound Healing
19.
Curr Opin Cell Biol ; 10(5): 667-71, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9818179

ABSTRACT

Analysis of extracellular matrix degradation systems has led to the insight that in cancer invasion there is often crucial interplay between cancer cells and several types of surrounding non-neoplastic stromal cells. Likewise, in normal tissue remodeling processes, the synthesis of proteolytic components is often distributed between several cell types, and there are strong similarities between neoplastic and non-neoplastic processes in the same tissue. Thus, tissue remodeling events are excellent models for studies of extracellular proteolysis in cancer. This has become even clearer by recent analyses of genetically modified mice.


Subject(s)
Extracellular Matrix Proteins/metabolism , Neoplasm Invasiveness , Regeneration , Breast Neoplasms/pathology , Colonic Neoplasms/pathology , Skin Neoplasms/pathology
20.
J Exp Med ; 188(6): 1029-37, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9743521

ABSTRACT

The urokinase receptor (CD87; uPAR) is found in close association with beta 2 integrins on leukocytes. We studied the functional consequence of this association for leukocyte adhesion and migration. In vivo, the beta 2 integrin-dependent recruitment of leukocytes to the inflamed peritoneum of uPAR-deficient mice was significantly reduced as compared with wild-type animals. In vitro, beta 2 integrin-mediated adhesion of leukocytes to endothelium was lost upon removal of uPAR from the leukocyte surface by phosphatidyl-inositol-specific phospholipase C. Leukocyte adhesion was reconstituted when soluble intact uPAR, but not a truncated form lacking the uPA-binding domain, was allowed to reassociate with the cell surface. uPAR ligation with a monoclonal antibody induced adhesion of monocytic cells and neutrophils to vascular endothelium by six- to eightfold, whereas ligation with inactivated uPA significantly reduced cell-to-cell adhesion irrespective of the beta 2 integrin-stimulating pathway. These data indicate that beta 2 integrin-mediated leukocyte-endothelial cell interactions and recruitment to inflamed areas require the presence of uPAR and define a new phenotype for uPAR-deficient mice. Moreover, uPAR ligation differentially modulates leukocyte adhesion to endothelium and provides novel targets for therapeutic strategies in inflammation-related vascular pathologies.


Subject(s)
CD18 Antigens/physiology , Cell Movement/immunology , Leukocytes/immunology , Plasminogen Activators/metabolism , Receptors, Cell Surface/physiology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CD18 Antigens/metabolism , Cell Adhesion/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Female , Humans , Leukocytes/enzymology , Leukocytes/metabolism , Mice , Mice, Knockout , Muscle, Smooth/cytology , Muscle, Smooth/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...