Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
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
2.
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
3.
Biochem Soc Trans ; 30(2): 177-83, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12023847

ABSTRACT

The ability to degrade the extracellular matrix by controlled proteolysis is an important property of malignant cancer cells, which enables them to invade the surrounding tissue and to gain access to the circulation by intravasation. One proteolytic system thought to be involved in these processes is urokinase-mediated plasminogen activation. Expression of a glycolipid-anchored receptor for urokinase-type plasminogen activator (uPA) targets this system to the cell surface. This receptor (uPAR) is composed of three homologous modules belonging to the Ly-6/uPAR/alpha-neurotoxin protein domain family. Integrity of the three-domain structure of uPAR is required for maintenance of its sub-nanomolar affinity for uPA, but the functional epitope for this interaction is primarily located in uPAR domain I. Using affinity maturation by combinatorial chemistry, we have recently identified a potent 9-mer peptide antagonist of the uPA-uPAR interaction having a high affinity for uPAR (K(d)< 1 nM). Photoaffinity labelling suggests that this peptide interacts with a composite binding site in uPAR involving both domains I and III. When tested in a chicken chorioallantoic membrane assay that was developed to quantify intravasation of human cells, this antagonist was able to reduce the intravasation of HEp-3 cancer cells by approx. 60%.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism , Amino Acid Sequence , Animals , Humans , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Molecular Structure , Neoplasm Invasiveness , Oligopeptides/chemistry , Oligopeptides/pharmacology , Receptors, Cell Surface/drug effects , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/drug effects
4.
Lab Invest ; 81(11): 1485-501, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11706057

ABSTRACT

Urokinase plasminogen activator (uPA) regulates a proteolytic cascade that facilitates cancer invasion through degradation of the extracellular matrix, and high levels of uPA in human breast cancer tissue correlate with poor prognosis. We previously found that, in ductal breast cancer, uPA mRNA is highly expressed by myofibroblasts surrounding invasively growing cancer cells. However, the localization of uPA protein has not been settled in the published literature. Because uPA is a secreted molecule, it could conceivably be localized differently from its mRNA. We have studied the localization of uPA immunoreactivity in detail. Twenty-five cases of invasive ductal carcinoma were analyzed with three different uPA antibody preparations, all of which gave an essentially identical stromal staining pattern. Using double immunofluorescence, we identified uPA immunoreactivity in myofibroblasts and macrophages in all cases examined. Additionally, in approximately half of the tumors, we saw uPA staining of endothelial cells. In 3 of the 25 cases, a small subpopulation of the cancer cells was uPA-positive. We conclude that uPA immunoreactivity is almost exclusively associated with stromal cells, which thus play a major role in generation of proteolytic activity in ductal breast cancer.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Stromal Cells/enzymology , Urokinase-Type Plasminogen Activator/analysis , Antibody Specificity , Biomarkers, Tumor , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Detergents , Enzyme-Linked Immunosorbent Assay , Female , Fixatives , Fluorescent Antibody Technique , Formaldehyde , Humans , In Situ Hybridization , Octoxynol , Paraffin Embedding , RNA, Messenger/analysis , Trypsin , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/immunology
5.
Biochemistry ; 40(40): 12157-68, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11580291

ABSTRACT

The high-affinity interaction between urokinase-type plasminogen activator (uPA) and its glycolipid-anchored receptor (uPAR) plays an important role in pericellular plasminogen activation. Since proteolytic degradation of the extracellular matrix has an established role in tumor invasion and metastasis, the uPA-uPAR interaction represents a potential target for therapeutic intervention. By affinity maturation using combinatorial chemistry we have now developed and characterized a 9-mer, linear peptide antagonist of the uPA-uPAR interaction demonstrating specific, high-affinity binding to human uPAR (K(d) approximately 0.4 nM). Studies by surface plasmon resonance reveal that the off-rate for this receptor-peptide complex is comparable to that measured for the natural protein ligand, uPA. The functional epitope on human uPAR for this antagonist has been delineated by site-directed mutagenesis, and its assignment to loop 3 of uPAR domain III (Met(246), His(249), His(251), and Phe(256)) corroborates data previously obtained by photoaffinity labeling and provides a molecular explanation for the extreme selectivity observed for the antagonist toward human compared to mouse, monkey, and hamster uPAR. When human HEp-3 cancer cells were inoculated in the presence of this peptide antagonist, a specific inhibition of cancer cell intravasation was observed in a chicken chorioallantoic membrane assay. These data imply that design of small organic molecules mimicking the binding determinants of this 9-mer peptide antagonist may have a potential application in combination therapy for certain types of cancer.


Subject(s)
Combinatorial Chemistry Techniques , Epitopes/chemistry , Neoplasms/pathology , Receptors, Cell Surface/antagonists & inhibitors , Animals , Base Sequence , Cell Line , Cricetinae , DNA Primers , Humans , Mice , Oligopeptides/chemistry , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
6.
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
7.
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
8.
Biochem J ; 358(Pt 3): 673-9, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11535128

ABSTRACT

Urokinase (uPA) has the striking ability to cleave its receptor, uPAR, thereby inactivating the binding potential of this molecule. Here we demonstrate that the glycosylphosphatidylinositol (GPI) anchor of uPAR, which is attached to the third domain, is an important determinant in governing this reaction, even though the actual cleavage occurs between the first and second domains. Purified full-length GPI-anchored uPAR (GPI-uPAR) proved much more susceptible to uPA-mediated cleavage than recombinant truncated soluble uPAR (suPAR), which lacks the glycolipid anchor. This was not a general difference in proteolytic susceptibility since GPI-uPAR and suPAR were cleaved with equal efficiency by plasmin. Since the amino acid sequences of GPI-uPAR and suPAR are identical except for the C-terminal truncation, the different cleavage patterns suggest that the two uPAR variants differ in the conformation or the flexibility of the linker region between domains 1 and 2. This was supported by the fact that an antibody to the peptide AVTYSRSRYLE, amino acids 84-94 in the linker region, recognizes GPI-uPAR but not suPAR. This difference in the linker region is thus caused by a difference in a remote hydrophobic region. In accordance with this model, when the hydrophobic lipid moiety was removed from the glycolipid anchor by phospholipase C, low concentrations of uPA could no longer cleave the modified GPI-uPAR and the reactivity to the peptide antibody was greatly decreased. Naturally occurring suPAR, purified from plasma, was found to have a similar resistance to uPA cleavage as phospholipase C-treated GPI-uPAR and recombinant suPAR.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , CHO Cells , Chromatography, Affinity , Cricetinae , Humans , Kinetics , Receptors, Cell Surface/isolation & purification , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection , U937 Cells
9.
Ugeskr Laeger ; 163(27): 3759-62, 2001 Jul 02.
Article in Danish | MEDLINE | ID: mdl-11466981

ABSTRACT

Our increased knowledge of basic molecular mechanisms involved in the growth and spreading of cancer has provided a new basis for development of anticancer drugs. As an example, matrix degrading protease systems are discussed, in particular the urokinase plasminogen activator (uPA) system in which inhibition of binding of uPA to its cellular receptor is an attractive target. However, biologically based drugs can in general be expected to present the same problems as conventional drugs, including toxic side effects due to the role of their targets under normal conditions, and development of resistance which among other mechanisms can be due to a widespread functional overlap, e.g. between different matrix degrading protease systems. Many still unexploited promising biological targets for cancer therapy are known and it is likely that a systematic, goal oriented research will result in many new drugs. These drugs may individually not necessarily be more effective than the conventional drugs, but they will, due to their different mechanisms of action, be well suited for use in combination with these.


Subject(s)
Antineoplastic Agents , Biopharmaceutics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Humans , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/metabolism , Models, Biological , Receptors, Drug/drug effects , Receptors, Drug/metabolism , Technology, Pharmaceutical , Urokinase-Type Plasminogen Activator/metabolism
10.
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
11.
Trends Cardiovasc Med ; 11(1): 7-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11413046

ABSTRACT

The urokinase-mediated plasminogen activation system plays a central role in the extracellular proteolytic degradation reactions in cancer invasion. In this review article we discuss a number of recent findings identifying a new cellular receptor protein, uPARAP, that interacts with components of this proteolytic system. uPARAP is a high molecular weight type-1 membrane protein, belonging to the macrophage mannose receptor protein family. On the surface of certain cells, uPARAP forms a ternary complex with the pro-form of the urokinase-type plasminogen activator (uPA) and its primary receptor (uPAR). While the biological consequences of this reaction have not yet been verified experimentally, a likely event is ligand internalization because uPARAP is a constitutively recycling internalization receptor. uPARAP also binds at least one component, collagen type V, in the extracellular matrix meshwork, pointing to a potential role in proteolytic substrate presentation. Additional ligands have been proposed, including collagenase-3 and glycoproteins capable of interacting with one of the multiple carbohydrate recognition-type domains of uPARAP. In various adult tissues uPARAP is present on fibroblasts, macrophages and a subset of endothelial cells. In fetal tissues the protein has also been demonstrated in certain bone forming regions. Hypotheses on the physiological function of uPARAP include regulatory roles in extracellular proteolysis. This type of function would be likely to direct the local turnover of proteases and their substrate degradation products and thus may add to the complicated interplay between several cell types in governing restricted tissue degradation.


Subject(s)
Mannose-Binding Lectins , Membrane Glycoproteins/physiology , Plasminogen Activators/physiology , Receptors, Cell Surface/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Humans , Membrane Glycoproteins/analysis , Membrane Glycoproteins/chemistry , Neoplasms/metabolism , Peptide Hydrolases/metabolism , Plasminogen Activators/chemistry , Protein Binding , Receptors, Cell Surface/analysis , Receptors, Cell Surface/chemistry , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/chemistry
12.
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
13.
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
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.
J Immunol Methods ; 235(1-2): 91-9, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10675761

ABSTRACT

A significant degree of nonspecificity was found in ELISA determinations of soluble urokinase receptor (suPAR) in human blood plasma when biotinylated monoclonal antibodies (Mabs) were used for the detection layer. Surface plasmon resonance studies using both nonbiotinylated and biotinylated antibodies demonstrated that biotinylation reduced specific binding of the antibodies to their target antigen, suPAR. Furthermore, biotinylation produced a new interaction with unknown human plasma protein(s), unrelated to suPAR. Nonspecific interaction with plasma protein(s) was also observed after biotinylation of a Mab having no specific target antigen in human plasma and, in both cases, the level of nonspecific interaction was directly related to the degree of antibody biotinylation. These results reinforce earlier observations that biotinylation of antibodies can reduce the affinity of antibodies, but also indicate that, in addition, biotinylation can reduce the specificity of immunoassays for plasma proteins.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , Biotinylation , Enzyme-Linked Immunosorbent Assay , Receptors, Cell Surface/blood , Humans , Receptors, Urokinase Plasminogen Activator , Surface Plasmon Resonance
16.
J Immunol Methods ; 234(1-2): 107-16, 2000 Feb 03.
Article in English | MEDLINE | ID: mdl-10669775

ABSTRACT

The urokinase receptor (uPAR) is a glycolipid anchored cell surface glycoprotein that plays a central role in extracellular proteolysis during tissue remodeling processes and cancer invasion. By intramuscular (i.m.) injection of rabbits with plasmid DNA coding for a carboxy-terminally truncated secreted form of the murine uPAR (muPAR), specific anti-sera with a titer of 64,000, as measured by ELISA, have been obtained. Rabbits received a total of 10 monthly injections of 1 mg DNA in phosphate-buffered saline. The antibody titer peaked between the 5th and 7th injection and slowly declined after the 8th injection. After the final immunization the immune response persisted for at least 6 months without further injections. The antibodies generated by DNA immunization were useful for immunohistochemistry and immunoblotting, recognizing the antigen both in its native and in its reduced and alkylated form. Using the antibodies in immunoblotting muPAR was identified in lysates of peritoneal macrophages, spleen and lung tissue. Both the intact and cleaved form of muPAR were identified in lysates of a murine monocyte cell line P388D.1. No cross-reaction with human uPAR was observed. In immunohistochemical analysis of normal mouse lung tissue uPAR immunoreactivity was located in the alveoli and pulmonary vessels, whereas the bronchial epithelium was negative. These results demonstrate that DNA immunization of rabbits using i.m. injection is a very effective and easy method to raise polyclonal antibodies which can be used for characterization and localization of muPAR in mouse tissue.


Subject(s)
Antibody Formation , Receptors, Cell Surface/genetics , Vaccines, DNA/immunology , Animals , Antibodies/immunology , Antibody Affinity , Cell Line , Gene Expression , Genetic Vectors , Humans , Immunoblotting , Immunohistochemistry/methods , Mice , Rabbits , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Staining and Labeling/methods , Time Factors
17.
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
18.
J Biol Chem ; 275(3): 1993-2002, 2000 Jan 21.
Article in English | MEDLINE | ID: mdl-10636902

ABSTRACT

The plasminogen activation cascade system, directed by urokinase and the urokinase receptor, plays a key role in extracellular proteolysis during tissue remodeling. To identify molecular interaction partners of these trigger proteins on the cell, we combined covalent protein cross-linking with mass spectrometry based methods for peptide mapping and primary structure analysis of electrophoretically isolated protein conjugates. A specific tri-molecular complex was observed upon addition of pro-urokinase to human U937 cells. This complex included the urokinase receptor, pro-urokinase, and an unknown, high molecular weight urokinase receptor-associated protein. The tryptic peptide mixture derived from a cross-linked complex of pro-urokinase and the latter protein was analyzed by nanoelectrospray tandem mass spectrometric sequencing. This analysis identified the novel protein as the human homologue of a murine membrane-bound lectin with hitherto unknown function. The human cDNA was cloned and sequenced. The protein, designated uPARAP, is a member of the macrophage mannose receptor protein family and contains a putative collagen-binding (fibronectin type II) domain in addition to 8 C-type carbohydrate recognition domains. It proved capable of binding strongly to a single type of collagen, collagen V. This collagen binding reaction at the exact site of plasminogen activation on the cell may lead to adhesive functions as well as a contribution to cellular degradation of collagen matrices.


Subject(s)
Collagen/metabolism , Mannose-Binding Lectins , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Cross-Linking Reagents/metabolism , Dose-Response Relationship, Drug , Glycosylation , Humans , Mass Spectrometry , Mice , Molecular Sequence Data , Muscle, Smooth, Vascular/metabolism , Plasminogen Activators/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Urokinase Plasminogen Activator , Sequence Homology, Amino Acid , U937 Cells
19.
Cancer Res ; 60(24): 6927-34, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11156392

ABSTRACT

We examined the relationship between tumor tissue level of the complex formed of urokinase (uPA) and its type-1 inhibitor (PAI-1) and survival of breast cancer patients. The study included 342 axillary lymph node-negative and -positive primary breast cancer patients with a median follow-up of 67 months. Using a newly established ELISA, the levels of preformed uPA-PAI-1 complex were measured in tumor tissue extracts and analyzed with respect to total uPA, total PAI-1, and clinicopathological parameters, including survival. uPA-PAI-1 complex comprised a minor, variable fraction of both total uPA and PAI-1 levels. The complex levels were higher in node-negative tumors than in node-positive tumors and higher in small and low-grade tumors (all, P < or = 0.002). The tumor levels of complex, uPA, and PAI-1 were all associated with survival; high complex levels predicted longer recurrence-free survival (P = 0.03) and overall survival [OS (P = 0.005)], whereas high uPA or PAI-1 levels significantly predicted shorter survival. In multivariate Cox analysis, the only parameters that independently predicted survival were total PAI-1 level and lymph node status for recurrence-free survival and OS and, additionally, steroid hormone receptor status and grade for OS. This is the first demonstration of a relationship between uPA.PAI-1 complex tumor level and patient survival. However, total PAI-1 level showed superior prognostic power. Additional studies are needed to understand the relationship of these parameters to cancer biology and to assess the clinical utility of the uPA PAI-1 complex.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Urokinase-Type Plasminogen Activator/metabolism , Adult , Aged , Blotting, Western , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lymph Nodes/metabolism , Lymphatic Metastasis , Middle Aged , Multivariate Analysis , Plasminogen Activator Inhibitor 1/biosynthesis , Time Factors , Urokinase-Type Plasminogen Activator/biosynthesis
20.
J Biol Chem ; 274(53): 37995-8003, 1999 Dec 31.
Article in English | MEDLINE | ID: mdl-10608868

ABSTRACT

The urokinase-type plasminogen activator receptor (uPAR) is a glycolipid anchored multidomain member of the Ly-6/uPAR protein domain superfamily. Studies by site-directed photoaffinity labeling, chemical cross-linking, and ligand-induced protection against chemical modification have highlighted the possible involvement of uPAR domain I and particularly loop 3 thereof in ligand binding (Ploug, M. (1998) Biochemistry 37, 16494-16505). Guided by these results we have now performed an alanine scanning analysis of this region in uPAR by site-directed mutagenesis and subsequently measured the effects thereof on the kinetics of uPA binding in real-time by surface plasmon resonance. Only four positions in loop 3 of uPAR domain I exhibited significant changes in the contribution to the free energy of uPA binding (DeltaDeltaG >/= 1.3 kcal mol(-1)) upon single-site substitutions to alanine (i.e. Arg(53), Leu(55), Tyr(57), and Leu(66)). The energetic impact of these four alanine substitutions was not caused by gross structural perturbations, since all monoclonal antibodies tested having conformation-dependent epitopes on this domain exhibited unaltered binding kinetics. These sites together with a three-dimensional structure for uPAR may provide an appropriate target for rational drug design aimed at developing new receptor binding antagonists with potential application in cancer therapy.


Subject(s)
Epitope Mapping , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Base Sequence , DNA Primers , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Thermodynamics , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...