Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Am Coll Cardiol ; 14(4): 837-49, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2677087

RESUMO

Thrombolytic therapy for the removal of intravascular thrombi was introduced when streptokinase was first given to humans 40 years ago, the same year the American College of Cardiology was founded. Streptokinase was first administered to patients with acute myocardial infarction in 1959. Today, thrombolytic therapy has been established to offer significant benefits to patients with acute myocardial infarction provided they are brought to medical attention early enough after the onset of symptoms. The two major agents, streptokinase and recombinant tissue-type plasminogen activator (rt-PA), have been shown to result in reperfusion of infarct-related arteries, to salvage ischemic myocardium, to improve myocardial performance and to reduce mortality. In spite of these impressive gains, this novel therapy has shortcomings. The interval from the start of thrombolytic treatment to coronary reperfusion varies significantly from patient to patient and may, at times, be too long to produce a real benefit in terms of salvage of ischemic myocardium. The rate of reocclusion lies somewhere between 10% and 20% and appears not to be influenced by concomitant heparin anticoagulation. The rate of bleeding complications even with the "fibrin-specific" rt-PA is higher than anticipated and may range from 10% to 30%. As a consequence, intensive efforts are being directed at the development of improved thrombolytic agents and for adjunctive therapy evaluating better anticoagulants than heparin and better antiplatelet agents than aspirin. This review is a status report summarizing where we are in thrombolytic therapy in acute myocardial infarction, where we need to improve treatment results and what is being done mainly at the preclinical level to bring about such improvements.


Assuntos
Fibrinolíticos/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Reperfusão Miocárdica , Terapia Trombolítica , Animais , Anticoagulantes/uso terapêutico , Aspirina/uso terapêutico , Heparina/uso terapêutico , Humanos , Inibidores da Agregação Plaquetária/uso terapêutico , Recidiva
2.
Cancer Gene Ther ; 7(2): 292-9, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10770639

RESUMO

The serine protease urokinase-type plasminogen activator, uPA, when bound to its specific receptor, uPAR (CD87), plays a significant role in tumor cell invasion and metastasis. In breast cancer, enhanced uPA antigen in the primary tumor is correlated with poor prognosis of the patient. In an in vivo nude mouse model, we tested tumor growth and metastasis of human breast carcinoma cells that had been transfected with an expression plasmid encoding a soluble form of uPAR (suPAR). We explored, whether suPAR/uPA interaction reduces the binding of uPA to cell surface-associated uPAR, and, as a consequence, could suppress tumor growth and metastasis of the human breast cancer cell line MDA-MB-231 BAG. Overexpressed, secreted suPAR was shown to bind and thus scavenge the uPA secreted by the transfected lines suPAR3 and suPAR10. In vitro, an overexpression of suPAR did not alter the proliferation rate of the transfected tumor cells, nor did it affect the expression of uPA. Overexpression of suPAR led to a reduction in the plasminogen activation-related proteolytic activity of breast carcinoma cells. Primary tumor growth in the mammary fat pad of nude mice was followed up for 52 days. Overexpression of suPAR correlated with a reduction in tumor growth (from day 21, reaching 30% by day 34) as well as lung colonization (lung metastasis-positive mice in suPAR3: 4 of 17; suPAR10: 3 of 10; parental MDA-MB-231 BAG: 13 of 18). We conclude that suPAR overexpression leading to effective scavenge of uPA impairs proteolysis as well as the tumor growth and metastatic potential of breast carcinoma cells in vivo.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Precursores Enzimáticos/biossíntese , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Receptores de Superfície Celular/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Células CHO , Divisão Celular/genética , Cricetinae , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Ligação Proteica , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Solubilidade , Transfecção , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
3.
Int J Oncol ; 13(4): 645-51, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9735390

RESUMO

The human ovarian cancer cell line OV-MZ-19, established from a patient with cystadenocarcinoma of the ovary, expressing thrombomodulin (TM), a cell surface receptor for the serine protease thrombin, interacts with monoclonal and polyclonal antibodies having different specificity for TM. These antibodies detect TM antigen by means of flow cytofluorometry, laser scanning microscopy, immunocytochemistry, and ELISA. Therefore a highly sensitive ELISA for TM antigen was established using two different monoclonal antibodies to quantify TM in tissue extracts and biological fluids, e.g. peritoneal malignant ascites. Primary malignant ovarian tumors and metastases of the omentum and intestine contain TM antigen as determined by ELISA but in significantly lower concentrations than benign ovarian tumors (p=0.0056). In contrast, malignant ascitic fluid of patients with advanced ovarian cancer (FIGO IIIc) contain significantly elevated concentrations of soluble TM than benign peritoneal exudates (p=0.0003). Immunoaffinity purified ascites-derived TM efficiently activates protein C. Protein C activation of ascites-derived TM as well as TM expressed by the tumor cells is inhibited by the monoclonal antibodies. TM abrogates the procoagulant activity of thrombin, reduces pericellular thrombin via internalization, accelerates the thrombin-mediated inactivation of pro-uPA, and the EGF domains of TM exhibit mitogenic activity towards fibroblasts and tumor cells. Both, thrombin and pro-uPA play important roles in tumor invasion and metastasis. Therefore, downregulation and/or release of TM into ascitic fluid may play an important role in the malignant behavior of tumor cells.


Assuntos
Líquido Ascítico/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/secundário , Receptores de Trombina/metabolismo , Trombomodulina/metabolismo , Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo/imunologia , Líquido Ascítico/química , Líquido Ascítico/patologia , Feminino , Humanos , Neoplasias Ovarianas/química , Neoplasias Ovarianas/metabolismo , Neoplasias Peritoneais/química , Neoplasias Peritoneais/metabolismo , Receptores de Superfície Celular/metabolismo , Serina Endopeptidases/metabolismo , Trombomodulina/imunologia , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/metabolismo
4.
Drug News Perspect ; 14(7): 401-11, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12813583

RESUMO

Invasiveness of a variety of tumors depends on the regulated expression of proteolytic enzymes that degrade the surrounding extracellular matrix and dissociate cell-cell and/or cell-matrix attachments. The tumor cell surface-associated urokinase-type plasminogen activator (uPA) system plays an especially important role in tumor cell invasion and metastasis. It consists of the serine protease uPA, its membrane-bound receptor (uPAR, CD87) and one of the natural inhibitors PAI-1 or PAI-2. There are strong indications based on animal experiments that interference with this system by inhibiting the enzymatic activity of uPA and/or antagonizing its binding to the receptor is of therapeutic relevance. With the recent solution of various X-ray structures of uPA/inhibitor complexes, structural information is available for optimizing existing lead compounds in their affinity and selectivity for uPA. Furthermore, peptide compounds capable of mimicking the structural epitope of uPA responsible for binding to the receptor efficiently antagonize this recognition process. Thus, both approaches prove to be well suited for the design of highly promising drugs in human medicine.

5.
Thromb Res ; 98(1): 73-81, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10706935

RESUMO

Plasminogen activator inhibitor type-1 is a key regulatory protein of the fibrinolytic system that is involved in a variety of physiological and pathophysiological processes. A panel of 14 monoclonal antibodies directed against plasminogen activator inhibitor type-1 was analyzed regarding epitope specificity on plasminogen activator inhibitor type-1. For this purpose, chimera consisting of plasminogen activator inhibitor type-1 and another plasminogen activator inhibitor, plasminogen activator inhibitor type-2, with different portions of the respective wild-type proteins, were generated and plasminogen activator inhibitor type-1-derived 20-mer and 10-mer linear peptides were synthesized. Nine of the 14 monoclonal antibodies recognized an epitope located in the region between amino acid 76-188 of plasminogen activator inhibitor type-1, which encompasses the binding sites for vitronectin, heparin, and part of the fibrin binding region. Of these nine monoclonal antibodies, six reacted with a quadruple plasminogen activator inhibitor type-1 mutant (N152H, K156T, Q321L, M356I), and seven detected a plasminogen activator inhibitor type-1 deletion mutant (DeltaF111-H114). Two of the remaining five monoclonal antibodies recognized epitopes located between amino acid 209-227 and amino acid 352-371, respectively, while the other three antibodies reacted with wild-type plasminogen activator inhibitor type-1, only. Additional experiments revealed that two of the 14 mAbs neutralized and one monoclonal antibodies increased plasminogen activator inhibitor type-1 activity toward urokinase-type plasminogen activator, one of its target proteases.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Mapeamento de Epitopos , Inibidor 1 de Ativador de Plasminogênio/imunologia , Humanos , Epitopos Imunodominantes , Fragmentos de Peptídeos/imunologia , Inibidor 2 de Ativador de Plasminogênio/imunologia , Proteínas Recombinantes de Fusão/imunologia
6.
Adv Exp Med Biol ; 477: 331-41, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10849761
7.
J Biol Chem ; 266(16): 10070-2, 1991 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-1645336

RESUMO

The kringle-2 domain of tissue plasminogen activator, cloned and expressed in Escherichia coli (Wilhelm, O.G., Jaskunas, S.R., Vlahos, C.J., and Bang, N.U. (1990) J. Biol. Chem. 265, 14606-14611), was internally radiolabeled using [35S]methionine-cysteine. Following refolding and isolation, the labeled polypeptide was further purified by reverse-phase high performance liquid chromatography. The purified kringle-2 domain was digested with thermolysin, and the resulting peptides were purified by high performance liquid chromatography. Five major peptides containing 35S were obtained. Amino acid sequence analysis showed that these peptides represented various cleavage products containing one or more of the following disulfides: Cys180-Cys261, Cys201-Cys243, Cys232-Cys256 (sequence numbering based on Pennica et al. (Pennica, D., Holmes, W.E., Kohr, W.J., Hakins, R.N., Vehar, G. A., Ward, C.A., Bennett, W.F., Yelverton E., Seeburg, P.H., Heynecker, H.L., Goeddel, E.V., and Collen, D. (1983) Nature 301, 214-221)). These results confirm that the refolding methodology used produced kringle-2 with the predicted disulfide linkage and, thus, yielded material suitable for structural and functional studies.


Assuntos
Dissulfetos/química , Escherichia coli/metabolismo , Fragmentos de Peptídeos/química , Ativador de Plasminogênio Tecidual/química , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão , Regulação Bacteriana da Expressão Gênica , Dados de Sequência Molecular , Mapeamento de Peptídeos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Termolisina , Ativador de Plasminogênio Tecidual/genética
8.
Exp Cell Res ; 224(2): 344-53, 1996 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8612711

RESUMO

We have previously demonstrated that vitronectin (VN), a morphoregulatory protein in the vessel wall, is internalized and translocated to the subendothelial matrix by an integrin-independent mechanism (J. Histochem. Cytochem. 41, 1823-1832, 1993). The cell surface component which mediates the initial contact of VN with endothelial cells is defined here. The specific binding of VN to endothelial cells demonstrated the following properties: a threefold increase after phorbol ester treatment; 85% inhibition by pretreatment of cells with phosphatidylinositol-phospholipase C to release glycolipid-anchored surface proteins; a 90% inhibition by urokinase (u-PA) receptor blocking antibody. u-PA increased VN binding to cells due to an eightfold increase in the affinity of VN for the u-PA receptor. Structure-function studies showed that the amino-terminal fragment of u-PA, devoid of any proteolytic activity, mediated this effect. Active plasminogen activator inhibitor-1 (PAI-1), but not inactivated PAI-1, inhibited VN binding to cells and displaced VN that was prebound to endothelial cell monolayers. Similarly, VN binding to purified (immobilized) u-PA receptor, but not to integrin, was enhanced by u-PA and inhibited by PAI-1. Hence, the binding of soluble VN to endothelial cell surfaces is mediated by the u-PA receptor, and the relative concentrations of u-PA and PAI-1 are able to regulate the strength of this interaction. Endothelial cell adhesion to immobilized VN was found to be integrin-mediated without any involvement of the VN-uPA-receptor system. Hence, the interaction of VN with the u-PA receptor may be involved in the regulation of cellular processes necessary for endothelial cell invasion and migration at VN-rich extracellular matrix sites.


Assuntos
Endotélio Vascular/química , Ativadores de Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Vitronectina/metabolismo , Especificidade de Anticorpos , Ligação Competitiva/fisiologia , Adesão Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Humanos , Radioisótopos do Iodo , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Ativadores de Plasminogênio/antagonistas & inibidores , Ligação Proteica/fisiologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais/fisiologia , Temperatura , Veias Umbilicais/citologia , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Vitronectina/imunologia
9.
J Biol Chem ; 265(24): 14606-11, 1990 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-2117612

RESUMO

The kringle-2 domain (residues 176-262) of tissue-type plasminogen activator (t-PA) was cloned and expressed in Escherichia coli. The recombinant peptide, which concentrated in cytoplasmic inclusion bodies, was isolated, solubilized, chemically refolded, and purified by affinity chromatography on lysine-Sepharose to apparent homogeneity. [35S]Cysteine-methionine-labeled polypeptide was used to study the interactions of kringle-2 with lysine, fibrin, and plasminogen activator inhibitor-1. The kringle-2 domain bound to lysine-Sepharose and to preformed fibrin with a Kd = 104 +/- 6.2 microM (0.86 +/- 0.012 binding site) and a Kd = 4.2 +/- 1.05 microM (0.80 +/- 0.081 binding site), respectively. Competition experiments and direct binding studies showed that the kringle-2 domain is required for the formation of the ternary t-PA-plasminogen-intact fibrin complex and that the association between the t-PA kringle-2 domain and fibrin does not require plasmin degradation of fibrin and exposure of new COOH-terminal lysine residues. We also observed that kringle-2 forms a complex with highly purified guanidine-activated plasminogen activator inhibitor-1, dissociable by 0.2 M epsilon-aminocaproic acid. The kringle-2 polypeptide significantly inhibited tissue plasminogen activator/plasminogen activator inhibitor-1 interaction. The kringle-2 domain bound to plasminogen activator inhibitor-1 in a specific and saturable manner with a Kd = 0.51 +/- 0.055 microM (0.35 +/- 0.026 binding site). Therefore, the t-PA kringle-2 domain is important for the interaction of t-PA not only with fibrin, but also with plasminogen activator inhibitor-1 and thus represents a key structure in the regulation of fibrinolysis.


Assuntos
Escherichia coli/genética , Ativador de Plasminogênio Tecidual/metabolismo , Clonagem Molecular , Fibrina/metabolismo , Humanos , Cinética , Lisina/metabolismo , Inativadores de Plasminogênio/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Radioisótopos de Enxofre , Ativador de Plasminogênio Tecidual/biossíntese , Ativador de Plasminogênio Tecidual/genética
10.
Proc Natl Acad Sci U S A ; 97(10): 5113-8, 2000 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-10805774

RESUMO

Increased expression of the serine protease urokinase-type plasminogen activator (uPA) in tumor tissues is highly correlated with tumor cell migration, invasion, proliferation, progression, and metastasis. Thus inhibition of uPA activity represents a promising target for antimetastatic therapy. So far, only the x-ray crystal structure of uPA inactivated by H-Glu-Gly-Arg-chloromethylketone has been reported, thus limited data are available for a rational structure-based design of uPA inhibitors. Taking into account the trypsin-like arginine specificity of uPA, (4-aminomethyl)phenylguanidine was selected as a potential P1 residue and iterative derivatization of its amino group with various hydrophobic residues, and structure-activity relationship-based optimization of the spacer in terms of hydrogen bond acceptor/donor properties led to N-(1-adamantyl)-N'-(4-guanidinobenzyl)urea as a highly selective nonpeptidic uPA inhibitor. The x-ray crystal structure of the uPA B-chain complexed with this inhibitor revealed a surprising binding mode consisting of the expected insertion of the phenylguanidine moiety into the S1 pocket, but with the adamantyl residue protruding toward the hydrophobic S1' enzyme subsite, thus exposing the ureido group to hydrogen-bonding interactions. Although in this enzyme-bound state the inhibitor is crossing the active site, interactions with the catalytic residues Ser-195 and His-57 are not observed, but their side chains are spatially displaced for steric reasons. Compared with other trypsin-like serine proteases, the S2 and S3/S4 pockets of uPA are reduced in size because of the 99-insertion loop. Therefore, the peculiar binding mode of the new type of uPA inhibitors offers the possibility of exploiting optimized interactions at the S1'/S2' subsites to further enhance selectivity and potency. Because crystals of the uPA/benzamidine complex allow inhibitor exchange by soaking procedures, the structure-based design of new generations of uPA inhibitors can rely on the assistance of x-ray analysis.


Assuntos
Guanidinas/química , Guanidinas/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/química , Sequência de Aminoácidos , Linhagem Celular , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Conformação Molecular , Dados de Sequência Molecular , Estrutura Molecular , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Relação Estrutura-Atividade
11.
Biol Chem ; 382(8): 1197-205, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11592401

RESUMO

Urokinase-type plasminogen activator (uPA) represents a central molecule in pericellular proteolysis and is implicated in a variety of physiological and pathophysiological processes such as tissue remodelling, wound healing, tumor invasion, and metastasis. uPA binds with high affinity to a specific cell surface receptor, uPAR (CD87), via a well defined sequence within the N-terminal region of uPA (uPA19-31). This interaction directs the proteolytic activity of uPA to the cell surface which represents an important step in tumor cell proliferation, invasion, and metastasis. Due to its fundamental role in these processes, the uPA/uPAR-system has emerged as a novel target for tumor therapy. Previously, we have identified a synthetic, cyclic, uPA-derived peptide, cyclo19,31uPA19-31, as a lead structure for the development of low molecular weight uPA-analogues, capable of blocking uPA/uPAR-interaction [Burgle et al., Biol. Chem. 378 (1997), 231-237]. We now searched for peptide variants of cyclo19,31uPA19-31 with elevated affinities for uPAR binding. Among other tasks, we performed a systematic D-amino acid scan of uPA19-31, in which each of the 13 L-amino acids was individually substituted by the corresponding D-amino acid. This led to the identification of cyclo19,31[D-Cys19]-uPA19-31 as a potent inhibitor of uPA/uPAR-interaction, displaying only a 20 to 40-fold lower binding capacity as compared to the naturally occurring uPAR-ligands uPA and its amino-terminal fragment. Cyclo19,31[D-Cys19]-uPA19-31 not only blocks binding of uPA to uPAR but is also capable of efficiently displacing uPAR-bound uPA from the cell surface and to inhibit uPA-mediated, tumor cell-associated plasminogen activation and fibrin degradation. Thus, cyclo19,31[D-Cys19]-uPA19-31 represents a promising therapeutic agent to significantly affect the tumor-associated uPA/uPAR-system.


Assuntos
Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/farmacologia , Receptores de Superfície Celular/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/síntese química , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Substituição de Aminoácidos , Ligação Competitiva , Membrana Celular/metabolismo , Células Cultivadas , Fibrina/metabolismo , Humanos , Concentração Inibidora 50 , Fragmentos de Peptídeos/metabolismo , Peptídeos Cíclicos/metabolismo , Plasminogênio/metabolismo , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Relação Estrutura-Atividade , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
12.
Biol Chem ; 382(5): 789-98, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11517932

RESUMO

Focussing of the serine protease urokinase (uPA) to the tumor cell surface via interaction with its receptor (uPAR) is an important step in tumor invasion and metastasis. The human ovarian cancer cell line OV-MZ-6#8 was stably transfected with expression plasmids either encoding cell-associated uPAR (GPI-uPAR) or a soluble form of uPAR (suPAR) lacking its glycan lipid anchor. In vitro, high level synthesis of functionally active recombinant suPAR inhibited cell proliferation and led to reduced cell-associated fibrin matrix degradation, whereas fibrinolytic activity was increased in OV-MZ-6#8 cells overexpressing GPI-uPAR. Both OV-MZ-6#8-derived clones were inoculated into the peritoneum of nude mice and tested for tumor growth and spread. High level synthesis of recombinant suPAR (without altering the physiological expression levels of GPI-uPAR and uPA in these cells) resulted in a significant reduction of tumor burden (up to 86%) in the xenogeneic mouse model. In contrast, overexpression of GPI-uPAR in tumor cells did not affect tumor growth. Our results demonstrate that high levels of suPAR in the ovarian cancer cell vicinity can act as a potent scavenger for uPA, thereby significantly reducing tumor cell growth and cancer progression in vivo.


Assuntos
Neoplasias Ovarianas/metabolismo , Neoplasias Peritoneais/patologia , Receptores de Superfície Celular/biossíntese , Animais , Divisão Celular , Feminino , Fibrinólise , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neoplasias Ovarianas/patologia , Fenótipo , Plasminogênio/metabolismo , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Solubilidade , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas/transplante
13.
J Cell Physiol ; 180(2): 225-35, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10395292

RESUMO

The glycosylphosphatidylinositol (GPI)-anchored, multifunctional receptor for the serine proteinase, urokinase plasminogen activator (uPAR, CD87), regulates plasminogen activation and cell migration, adhesion, and proliferation. uPAR occurs in functionally distinct, membrane-anchored and soluble isoforms (s-uPAR) in vitro and in vivo. Recent evidence indicates that s-uPAR present in the circulation of cancer patients correlates with tumor malignancy and represents a valuable prognostic marker in certain types of cancer. We have therefore analyzed the mechanism of uPAR shedding in vitro. We present evidence that uPAR is actively released from ovarian cancer cells since the rate of receptor shedding did not correlate with uPAR expression. While s-uPAR was derived from the cell surface, it lacked the hydrophobic portion of the GPI moiety indicating anchor cleavage. We show that uPAR release is catalyzed by cellular GPI-specific phospholipase D (GPI-PLD), an enzyme cleaving the GPI anchor of the receptor. Thus, recombinant GPI-PLD expression increased receptor release up to fourfold. Conversely, a 40% reduction in GPI-PLD activity by GPI-PLD antisense mRNA expression inhibited uPAR release by more than 60%. We found that GPI-PLD also regulated uPAR expression, possibly by releasing a GPI-anchored growth factor. Our data suggest that cellular GPI-PLD might be involved in the generation of circulating prognostic markers in cancer and possibly regulate the function of GPI-anchored proteins by generating functionally distinct, soluble counterparts.


Assuntos
Glicosilfosfatidilinositóis/metabolismo , Fosfolipase D/metabolismo , Ativadores de Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Elementos Antissenso (Genética) , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glicosilfosfatidilinositóis/genética , Humanos , Hidrólise , Proteínas de Membrana/metabolismo , Neoplasias Ovarianas , Fosfolipase D/genética , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA