Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Thromb Haemost ; 100(6): 1068-75, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19132232

RESUMO

During systemic inflammation, neutrophil activation is accompanied by endothelial cell damage and hypercoagulability. Activated neutrophils release serine proteases that participate in tissue injury. We sought to investigate the effects of neutrophil proteases on proinflammatory and procoagulant changes in endothelial cells. The effects of elastase (HNE), cathepsin G (CG), and proteinase 3 (PR3) on expression of tissue factor (TF) and tissue factor pathway inhibitor-1 (TFPI) were examined in human umbilical vein endothelial cells. Flow cytometry demonstrated that these proteases proteolytically degraded endothelial cell-bound TFPI. TFPI mRNA expression was reduced by HNE and CG. PR3, but not HNE or CG, increased surface expression of TF and TF mRNA. Yet, increased TF expression did not enhance TF activity suggesting induction of encrypted TF. Using antibodies and siRNA to inhibit and silence PAR-1 and PAR-2, we observed that PR3 upregulation of TF is at least in part mediated by PAR-1. Although CG and HNE cleaved PAR-1, antibody reactivity to the PAR-1 hirudin-like sequence demonstrated inactivating cleavage, accounting for the selective ability of PR3 to induce PAR-1-mediated procoagulant effects. This was supported by induction of p42/44 MAPK by PR3. In conclusion, PR3 degradation of TFPI increases the procoagulant activity of endothelial cells. Release of PR3 after neutrophil activation may represent an important step in neutrophil-mediated vascular injury.


Assuntos
Coagulação Sanguínea , Catepsinas/metabolismo , Células Endoteliais/enzimologia , Elastase de Leucócito/metabolismo , Lipoproteínas/metabolismo , Mieloblastina/metabolismo , Neutrófilos/enzimologia , Serina Endopeptidases/metabolismo , Tromboplastina/metabolismo , Catepsina G , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Citometria de Fluxo , Humanos , Lipoproteínas/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ativação de Neutrófilo , Fragmentos de Peptídeos/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor PAR-1/agonistas , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Fatores de Tempo
2.
Arterioscler Thromb Vasc Biol ; 27(4): 769-75, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17255532

RESUMO

OBJECTIVE: The serine protease MT-SP1/matriptase plays an important role in cell migration and matrix degradation. Hepatocyte growth factor (HGF), urokinase-type plasminogen activator (uPA), and protease-activated receptor 2 (PAR-2) have been identified as in vitro substrates of MT-SP1/matriptase. Because PAR-2 is expressed in endothelial cells and contributes to inflammatory processes, we sought to investigate the effects of MT-SP1/matriptase on endothelial cytokine expression and analyzed MT-SP1/matriptase expression in vascular cells and atherosclerotic lesions. METHODS AND RESULTS: In endothelial cells, recombinant MT-SP1/matriptase dose-dependently induced interleukin (IL)-8 and IL-6 mRNA and protein expression dependent on its proteolytic activity. MT-SP1/matriptase time-dependently induced phosphorylation of p38 MAPK and p42/44 MAPK. Inhibitor experiments revealed that p38 MAPK and PKCalpha were necessary for IL-8 induction. PAR-2 downregulation abolished and PAR-2 overexpression augmented MT-SP1/matriptase-induced IL-8 expression as evidence for PAR-2 signaling. In human atherectomies, MT-SP1/matriptase was expressed in blood cells adherent to the endothelium. Concordantly, basal MT-SP1/matriptase expression was detected in isolated monocytes. Coincubation of monocytes and endothelial cells resulted in an increased IL-8 release, which was reduced after downregulation of endothelial PAR-2 and monocytic MT-SP1/matriptase. CONCLUSION: MT-SP1/matriptase induces release of proinflammatory cytokines in endothelial cells through activation of PAR-2. MT-SP1/matriptase is expressed in monocytes, thus, interaction of monocytic MT-SP1/matriptase with endothelial PAR-2 may contribute to atherosclerosis.


Assuntos
Células Endoteliais/metabolismo , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Receptor PAR-2/metabolismo , Serina Endopeptidases/farmacologia , Aterosclerose/enzimologia , Aterosclerose/etiologia , Catálise , Células Cultivadas , Citocinas/biossíntese , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Humanos , Interleucina-6/genética , Interleucina-8/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monócitos/metabolismo , NF-kappa B/metabolismo , Proteína Quinase C-alfa/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Serina Endopeptidases/sangue , Serina Endopeptidases/isolamento & purificação , Serina Endopeptidases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Circ Res ; 97(3): 293-8, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-16020755

RESUMO

Tissue factor (TF), the cell surface receptor for the serine protease FVIIa supports cell migration by interaction with the cytoskeleton. Intracellular signaling pathways dependent on the cytoplasmic domain of TF modify cell migration and may alter vascular remodeling. Vascular remodeling was analyzed in a femoral artery injury and a blood flow cessation model in mice with a targeted deletion of the 18 carboxy-terminal intracellular amino acids of TF (TF(Deltact/Deltact)) and compared with TF wild-type mice (TF(wt/wt)). Morphometric analysis revealed a decrease in the intima/media ratio after vascular injury in arteries from TF(Deltact/Deltact) compared with TF(wt/wt) mice (femoral artery injury: 2.4+/-0.3 TF(wt/wt) versus 0.6+/-0.3 TF(Deltact/Deltact), n=9 to 10, P=0.002; carotis ligation: 0.45+0.11 TF(wt/wt) versus 0.22+0.03 TF(Deltact/Deltact), n=12 to 14, P=0.09). This was caused by an increase in the media by 54% (P=0.04) in the femoral artery model and by 32% (P=0.03) after carotis ligation and was associated with an increased number of proliferating cells. Isolated aortic smooth muscle cells (SMCs) of TF(wt/wt) mice showed an increased migratory response toward the TF ligand active site-inhibited FVIIa that was abolished in TF(Deltact/Deltact) SMC. In contrast, the unstimulated proliferation rate was increased in TF(Deltact/Deltact) SMC compared with TF(wt/wt) SMCs. Thus, retention of SMCs attributable to a migratory defect and increased proliferation results in thickening of the media and in decrease in neointima formation after arterial injury. TF cytoplasmic domain signaling alters vascular remodeling and, thereby, may play a role in the development of restenosis, atherosclerotic disease, and neovascularization.


Assuntos
Citoplasma/química , Tromboplastina/fisiologia , Túnica Íntima/patologia , Túnica Média/patologia , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Artéria Femoral/patologia , Camundongos , Músculo Liso Vascular/patologia , Tromboplastina/química
4.
Circulation ; 111(3): 349-55, 2005 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-15642762

RESUMO

BACKGROUND: Tissue factor (TF), the surface receptor for the serine protease factor VIIa (FVIIa) and the initiator of the extrinsic coagulation cascade, supports vessel development and tumor metastasis by activation of extracellular, protease-dependent signaling pathways. The molecular mechanisms that do not require proteolytic activity of FVIIa are not yet known. The aim of the study, therefore, was to investigate the effects of active-site-inhibited FVIIa (FFR-FVIIa) on TF-mediated signaling. METHODS AND RESULTS: After stimulation with FVIIa and FFR-FVIIa, migration and activation of the GTPase Rac (Rac1) or the mitogen-activated protein kinase p38 (p38) were analyzed in J82 cells. FVIIa and FFR-FVIIa stimulated migration and activation of Rac1 and p38 in a TF-specific, dose- and time-dependent manner. Enhancement of migration required activation of Rac1 and p38, because it was abolished after inhibition with SB203580 or overexpression of dominant negative p38 and Rac1. The cytoplasmic domain of TF was necessary because no effects of FFR-FVIIa could be detected after transfection of a TF deletion mutant lacking the cytoplasmic domain. CONCLUSIONS: We identified a novel signaling pathway through which TF stimulates migration by activation of p38 and Rac1 independent of the proteolytic activity of FVIIa but dependent on the cytoplasmic domain of TF. Binding of FFR-VIIa to TF may stimulate vessel wall remodeling by enhancement of migration through activation of Rac1 and p38. This novel link may provide an insight into the understanding of the nonhemostatic functions of TF.


Assuntos
Tromboplastina/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Citoplasma/metabolismo , Ativação Enzimática , Fator VIIa/farmacologia , Fator VIIa/fisiologia , Humanos , Estrutura Terciária de Proteína , Transdução de Sinais
5.
Arterioscler Thromb Vasc Biol ; 25(2): 461-6, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15550696

RESUMO

OBJECTIVE: In acute myocardial infarction (AMI), proinflammatory plasma C-reactive protein values are strongly associated with postinfarction morbidity and mortality. So far, the cause of these inflammatory changes is not well understood. Therefore, we sought to investigate the relationship between the activation of coagulation and subsequent systemic inflammatory changes in AMI. METHODS AND RESULTS: Factor Xa (FXa) bound to tissue factor pathway inhibitor and prothrombin fragments F1+2 (F1+2) were used as a measure for activated coagulation. To assess systemic inflammatory changes, plasma interleukin (IL)-6 and IL-8 concentrations were analyzed by immunoassay. Blood samples were taken from 21 patients with AMI and 20 patients with stable angina pectoris. In AMI, tissue factor pathway inhibitor FXa but not F1+2 plasma levels were associated with circulating IL-8 (P=0.01). In vitro experiments revealed that FXa stimulated IL-8 and monocyte chemoattractant protein-1 release and RNA expression in endothelial cells and mononuclear leukocytes by activation of protease-activated receptor-1. CONCLUSIONS: Our data suggest that coagulation FXa may contribute to proinflammatory changes in AMI by stimulation of IL-8 release. Therapeutic inhibition of the proinflammatory effects of FXa may improve the clinical course in AMI. This study investigates the relationship between the activation of coagulation and systemic inflammatory changes in acute myocardial infarction. Tissue factor pathway inhibitor factor Xa but not F1+2 plasma levels were associated with circulating interleukin-8. In vitro factor Xa stimulated interleukin-8 and monocyte chemoattractant protein-1 release and RNA expression by activation of protease-activated receptor 1 as an underlying mechanism.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Fator Xa/fisiologia , Interleucina-8/metabolismo , Leucócitos Mononucleares/metabolismo , Infarto do Miocárdio/sangue , Adulto , Idoso , Angina Pectoris/sangue , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Quimiocina CCL2/metabolismo , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Fator Xa/farmacologia , Inibidores do Fator Xa , Feminino , Hirudinas/farmacologia , Humanos , Inflamação/sangue , Interleucina-6/sangue , Interleucina-8/sangue , Leucócitos Mononucleares/efeitos dos fármacos , Lipoproteínas/análise , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/farmacologia , Protrombina/análise , Proteínas Recombinantes/farmacologia , Veias Umbilicais
6.
J Biol Chem ; 279(28): 28989-97, 2004 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-15123706

RESUMO

Negative regulation of mitogenic pathways is a fundamental process that remains poorly characterized. The angiotensin II AT2 receptor is a rare example of a 7-transmembrane domain receptor that negatively cross-talks with receptor tyrosine kinases to inhibit cell growth. In the present study, we report the molecular cloning of a novel protein, ATIP1 (AT2-interacting protein), which interacts with the C-terminal tail of the AT2 receptor, but not with those of other receptors such as angiotensin AT1, bradykinin BK2, and adrenergic beta(2) receptor. ATIP1 defines a family of at least four members that possess the same domain of interaction with the AT2 receptor, contain a large coiled-coil region, and are able to dimerize. Ectopic expression of ATIP1 in eukaryotic cells leads to inhibition of insulin, basic fibroblast growth factor, and epidermal growth factor-induced ERK2 activation and DNA synthesis, and attenuates insulin receptor autophosphorylation, in the same way as the AT2 receptor. The inhibitory effect of ATIP1 requires expression, but not ligand activation, of the AT2 receptor and is further increased in the presence of Ang II, indicating that ATIP1 cooperates with AT2 to transinactivate receptor tyrosine kinases. Our findings therefore identify ATIP1 as a novel early component of growth inhibitory signaling cascade.


Assuntos
Proteínas de Transporte/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Transporte/genética , Divisão Celular/fisiologia , Chlorocebus aethiops , Clonagem Molecular , Dimerização , Ativação Enzimática , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Insulina/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptor Tipo 2 de Angiotensina/química , Receptor Tipo 2 de Angiotensina/genética , Alinhamento de Sequência , Distribuição Tecidual , Proteínas Supressoras de Tumor/genética , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...