Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 31(12): 5419-5431, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28808141

RESUMO

The plasma kallikrein-kinin system (KKS) consists of serine proteases, prekallikrein (pKal) and factor XII (FXII), and a cofactor, high-MW kininogen (HK). Upon activation, activated pKal and FXII cleave HK to release bradykinin. Activation of this system has been noted in patients with rheumatoid arthritis, and its pathogenic role has been characterized in animal arthritic models. In this study, we generated 2 knockout mouse strains that lacked pKal and HK and determined the role of KKS in autoantibody-induced arthritis. In a K/BxN serum transfer-induced arthritis (STIA) model, mice that lacked HK, pKal, or bradykinin receptors displayed protective phenotypes in joint swelling, histologic changes in inflammation, and cytokine production; however, FXII-deficient mice developed normal arthritis. Inhibition of Kal ameliorated arthritis severity and incidence at early stage STIA and reduced the levels of major cytokines in joints. In addition to releasing bradykinin from HK, Kal directly activated monocytes to produce proinflammatory cytokines, up-regulated their C5aR and FcRIII expression, and released C5a. Immune complex increased pKal activity, which led to HK cleavage. The absence of HK is associated with a decrease in joint vasopermeability. Thus, we identify a critical role for Kal in autoantibody-induced arthritis with pleiotropic effects, which suggests that it is a new target for the inhibition of arthritis.-Yang, A., Zhou, J., Wang, B., Dai, J., Colman, R. W., Song, W., Wu, Y. A critical role for plasma kallikrein in the pathogenesis of autoantibody-induced arthritis.


Assuntos
Artrite/metabolismo , Artrite/patologia , Autoanticorpos/metabolismo , Calicreína Plasmática/metabolismo , Animais , Artrite/genética , Artrite/imunologia , Bradicinina/metabolismo , Citocinas/metabolismo , Fator XII/genética , Fator XII/metabolismo , Feminino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Calicreína Plasmática/genética , Reação em Cadeia da Polimerase
2.
J Immunol ; 192(9): 4398-408, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24688027

RESUMO

Phagocytosis of apoptotic cells (efferocytosis) is essential for regulation of immune responses and tissue homeostasis and is mediated by phagocytic receptors. In this study, we found that urokinase plasminogen activator receptor (uPAR) plays an important role in internalization of apoptotic cells and also characterized the underlying mechanisms. In a flow cytometry-based phagocytic assay, uPAR-deficient macrophages displayed significant defect in internalization but not tethering of apoptotic cells. When uPAR-deficient mice were challenged with apoptotic cells, they exhibited pronounced splenomegaly resulting from accumulation of abundant apoptotic cells in spleen. Overexpression of uPAR in HEK-293 cells enhanced efferocytosis, which was inhibited by Annexin V and phosphatidylserine (PS) liposome, suggesting that uPAR-mediated efferocytosis is dependent on PS. In serum lacking high m.w. kininogen (HK), a uPAR ligand, uPAR-mediated efferocytosis was significantly attenuated, which was rescued by replenishment of HK. As detected by flow cytometry, HK selectively bound to apoptotic cells, but not viable cells. In purified systems, HK was specifically associated with PS liposome. HK binding to apoptotic cells induced its rapid cleavage to the two-chain form of HK (HKa) and bradykinin. Both the H chain and L chain of HKa were associated with PS liposome and apoptotic cells. HKa has higher binding affinity than HK to uPAR. Overexpression of Rac1/N17 cDNA inhibited uPAR-mediated efferocytosis. HK plus PS liposome stimulated a complex formation of CrkII with p130Cas and Dock-180 and Rac1 activation in uPAR-293 cells, but not in control HEK-293 cells. Thus, uPAR mediates efferocytosis through HK interaction with PS on apoptotic cells and activation of the Rac1 pathway.


Assuntos
Macrófagos/imunologia , Fagocitose/fisiologia , Fosfatidilserinas/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Apoptose/imunologia , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Cininogênio de Alto Peso Molecular/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia , Ressonância de Plasmônio de Superfície , Transfecção , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
Arthritis Rheum ; 64(11): 3574-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22739815

RESUMO

OBJECTIVE: To examine whether activation of the plasma kallikrein-kinin system (KKS) mediates synovial recruitment of endothelial progenitor cells (EPCs) in arthritis. METHODS: EPCs were isolated from Lewis rat bone marrow, and expression of progenitor cell-lineage markers and functional properties were characterized. EPCs were injected intravenously into Lewis rats with arthritis, and their recruitment and formation of de novo blood vessels in inflamed synovium were evaluated. The role of plasma KKS was examined using a plasma kallikrein inhibitor (EPI-KAL2) and an antikallikrein antibody (13G11). A transendothelial migration assay was used to determine the role of bradykinin and its receptor in EPC mobilization. RESULTS: EPCs from Lewis rats exhibited a strong capacity to form tubes and vacuoles and expressed increased levels of bradykinin type 2 receptor (B2R) and progenitor cell markers CD34 and Sca-1. In Lewis rats with arthritis, EPCs were recruited into inflamed synovium at the acute phase of disease and formed de novo blood vessels. Inhibition of plasma kallikrein by EPI-KAL2 and 13G11 significantly suppressed synovial recruitment of EPCs and hyperproliferation of synovial cells. Bradykinin stimulated transendothelial migration of EPCs in a concentration-dependent manner. This was mediated by B2R, as demonstrated by the finding that knockdown of B2R with silencing RNA completely blocked bradykinin-stimulated transendothelial migration. Moreover, bradykinin selectively up-regulated expression of the homing receptor CXCR4 in EPCs. CONCLUSION: These observations demonstrate a novel role of plasma KKS activation in the synovial recruitment of EPCs in arthritis, acting via kallikrein activation and B2R-dependent mechanisms. B2R might be involved in the mobilization of EPCs via up-regulation of CXCR4.


Assuntos
Artrite Experimental/imunologia , Endotélio Vascular/imunologia , Células-Tronco Hematopoéticas/imunologia , Sistema Calicreína-Cinina/imunologia , Receptor B2 da Bradicinina/imunologia , Membrana Sinovial/imunologia , Doença Aguda , Animais , Artrite Experimental/patologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Bradicinina/imunologia , Movimento Celular/imunologia , Células Cultivadas , Endotélio Vascular/citologia , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Calicreínas/sangue , Calicreínas/imunologia , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Ratos Transgênicos , Receptores CXCR4/genética , Receptores CXCR4/imunologia , Membrana Sinovial/irrigação sanguínea , Membrana Sinovial/patologia
4.
J Exp Med ; 203(3): 493-5, 2006 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-16533890

RESUMO

Factor XII (FXII), a clotting enzyme that can initiate coagulation in vitro, has long been considered dispensable for normal blood clotting in vivo because hereditary deficiencies in FXII are not associated with spontaneous or excessive bleeding. However, new studies show that mice lacking FXII are protected against arterial thrombosis (obstructive clot formation) and stroke. Thus, FXII could be a unique drug target that could be blocked to prevent thrombosis without the side effect of increased bleeding.


Assuntos
Isquemia Encefálica/metabolismo , Deficiência do Fator XII/metabolismo , Hemostasia , Trombose/metabolismo , Animais , Inibidores dos Fatores de Coagulação Sanguínea/administração & dosagem , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Deficiência do Fator XI/tratamento farmacológico , Deficiência do Fator XI/metabolismo , Deficiência do Fator XI/patologia , Fator XII/antagonistas & inibidores , Fator XII/metabolismo , Deficiência do Fator XII/patologia , Feminino , Fibrina/metabolismo , Hemostasia/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Trombose/tratamento farmacológico , Trombose/patologia
5.
Arterioscler Thromb Vasc Biol ; 31(4): 883-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21252071

RESUMO

OBJECTIVE: Cleaved high-molecular-weight kininogen (HKa), an activation product of the plasma kallikrein-kinin system, inhibits endothelial cell functions. We questioned whether HKa affects the function of endothelial progenitor cells (EPCs) and accelerates their senescence. METHODS AND RESULTS: Treatment with HKa for 2 weeks markedly inhibited the formation of large colonies and proliferation of EPCs on collagen surfaces, whereas HKa did not affect collagen-mediated EPC adhesion and survival. Concomitantly, treated EPCs displayed flattened and giant cell morphological changes and formation of intracellular vacuoles. As determined by acidic ß-galactosidase staining, HKa increased senescent EPCs by 2- and >3-fold after culture for 1 and 2 weeks, respectively. In addition, HKa suppressed the telomerase activity of EPCs. HKa concentration-dependently increased the generation of intracellular reactive oxygen species (ROS) and markedly upregulated p38 kinase phosphorylation and prosenescence molecule p16(INK4a) expression. SB203580, a p38 inhibitor, attenuated the level of HKa-enhanced p16(INK4a) expression. Either quenching of ROS or inhibition of p38 kinase prevented HKa-induced EPC senescence. CONCLUSIONS: HKa accelerates the onset of EPC senescence by activating the ROS-p38 kinase-p16(INK4a) signaling cascade. This novel activity of HKa points out the likelihood of HKa serving as an endogenous inducer of EPC senescence.


Assuntos
Proliferação de Células , Senescência Celular , Células Endoteliais/metabolismo , Cininogênio de Alto Peso Molecular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Adesão Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Colágeno/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Endoteliais/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Células-Tronco/efeitos dos fármacos , Telomerase/metabolismo , Fatores de Tempo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 298(2): H652-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19966052

RESUMO

Inflammatory bowel disease and arthritis are associated with contact activation that results in cleavage of kininogen to form high molecular weight kininogen (HKa) and bradykinin. We have previously demonstrated that HKa can stimulate inflammatory cytokine and chemokine secretion from human monocytes. We now show that HKa can upregulate tissue factor antigen and procoagulant activity on human monocytes as a function of time (1-4 h) and HKa concentration (75-900 nM). The amino acid sequence responsible to block HKa effects is G440-H455. The HKa receptor macrophage-1 (Mac-1; CD11b18) is the binding site as shown by inhibition by a monoclonal antibody to CD11b/18. Chemical inhibitors of JNK, ERK, and p38 signaling pathways block cell signaling, as does an inhibitor to the transcription factor NF-kappaB. A combination of monoclonal antibodies to TNF-alpha and IL-1beta but neither alone inhibited the HKa induction of tissue factor. These results suggest that HKa mimics LPS by triggering a paracrine pathway in monocytes that depends on TNF-alpha and IL-1beta. Antibodies to kininogen or peptidomimetics might be a useful and safe therapy in inflammatory diseases or sepsis involving cytokines.


Assuntos
Interleucina-1beta/metabolismo , Cininogênio de Alto Peso Molecular/farmacologia , Monócitos/metabolismo , Tromboplastina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/imunologia , Cininogênio de Alto Peso Molecular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Tromboplastina/genética , Fatores de Tempo , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/fisiologia
7.
Cancer Immunol Immunother ; 59(12): 1885-93, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20811885

RESUMO

Metastasis of malignant tumors is a major cause of morbidity and mortality. Inhibition of tumor growth in distant organs is of clinical importance. We have demonstrated that C11C1, a murine monoclonal antibody to the light chain region of high molecular weight kininogen (HK), reduces growth of murine multiple myeloma in normal mice and human colon cancer in nude mice. C11C1 inhibits angiogenesis by reducing tumor microvascular density by blocking binding of HK to endothelial cells. We now evaluate the anti-metastatic effect of C11C1 on C57BL/6 mouse lung metastatic model using B16F10 melanoma cells. The tail veins of mice were injected with 0.5 × 10(6) cells of melanoma B16F10. One group received C11C1 and the other received saline (control) intraperitoneally. When mice were killed at 28 days, 6 of 10 control mice had detectable metastatic pulmonary nodules which stained positive with an antibody against S-100 protein, a tumor antigen present in malignant melanoma cells. In the C11C1 groups, none of the mice showed metastatic foci in their lungs. We showed that C11C1 inhibits endothelial cell tube formation in a 3-D collagen fibrinogen gel model by inhibiting the rate of cleavage of HK by plasma kallikrein without changing the binding affinity for HK. These studies demonstrate that a monoclonal antibody to HK has the potential to prevent metastasis with minimal side effects.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Cininogênio de Alto Peso Molecular/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Cininogênio de Alto Peso Molecular/metabolismo , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína
8.
Arterioscler Thromb Vasc Biol ; 27(9): 1968-75, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17585065

RESUMO

OBJECTIVE: The cleaved form of high molecular weight kininogen (HKa) is a potent inhibitor of angiogenesis and tumor growth in vivo; the functional domain has been identified as domain 5 (D5, named as kininostatin). We now identify the subcellular targeting site for D5 on endothelial cells (ECs), and investigate D5 inhibition of integrin functions. METHODS AND RESULTS: Endothelial membrane rafts were isolated using sucrose density gradient centrifugation. D5, bound to ECs, was predominantly associated with membrane rafts, in which uPAR, a HKa receptor, was also localized. In contrast, other HKa receptors, cytokeratin-1 and gC1q receptor, were not detected in membrane rafts. Colocalization of D5 with caveolin-1 was demonstrated on ECs by confocal microscopy. Disruption of membrane rafts by cholesterol removal decreased D5 binding to ECs. On stimulation with vascular endothelial growth factor, alpha(v)beta3 integrin formed a complex with uPAR and caveolin-1, which was accompanied by an increase in ligand binding affinity of alpha(v)beta3 integrin. These events were inhibited by D5. Consistently, D5 suppressed specific alpha(v)beta3 integrin-mediated EC adhesion and spreading as well as small guanosine triphosphatase Rac1 activation. CONCLUSIONS: D5 binds to ECs via membrane rafts and downregulates alpha(v)beta3 integrin bidirectional signaling and the downstream Rac1 activation pathway.


Assuntos
Adesão Celular/fisiologia , Células Endoteliais/fisiologia , Integrina alfaVbeta3/metabolismo , Cininogênio de Alto Peso Molecular/fisiologia , Inibidores da Angiogênese/fisiologia , Membrana Celular/metabolismo , Regulação para Baixo , Humanos , Veias Umbilicais/fisiologia
9.
Bioorg Chem ; 36(3): 141-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18394675

RESUMO

The amino acids involved in substrate (cAMP) binding to human platelet cGMP-inhibited cAMP phosphodiesterase (PDE3A) are identified. Less is known about the inhibitor (cGMP) binding site. We have now synthesized a nonhydrolyzable reactive cGMP analog, Rp-guanosine-3',5'-cyclic-S-(4-bromo-2, 3-dioxobutyl)monophosphorothioate (Rp-cGMPS-BDB). Rp-cGMPS-BDB irreversibly inactivates PDE3A (K(I)=43.4+/-7.2muM and k(cart)=0.007+/-0.0006 min(-1)). The effectiveness of protectants in decreasing the rate of inactivation by Rp-cGMPS-BDB is: Rp-cGMPS (K(d)=72 microM)>Sp-cGMPS (124), Sp-cAMPS (182)>GMP (1517), Rp-cAMPS (3762), AMP (4370 microM). NAD(+), neither a substrate nor an inhibitor of PDE3A, does not protect. Nonhydrolyzable cGMP analogs exhibit greater affinity than the cAMP analogs. These results indicate that Rp-cGMPS-BDB targets favorably the cGMP binding site consistent with a docking model of PDE3A-Rp-cGMPS-BDB active site. We conclude that Rp-cGMPS-BDB is an effective active site-directed affinity label for PDE3A with potential for other cGMP-dependent enzymes.


Assuntos
Marcadores de Afinidade/química , GMP Cíclico/análogos & derivados , Inibidores Enzimáticos/química , Inibidores da Fosfodiesterase 3 , Marcadores de Afinidade/síntese química , Sítios de Ligação , Plaquetas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Ligação Proteica
10.
Front Immunol ; 9: 21, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29467753

RESUMO

The kallikrein-kinin system (KKS) consists of two serine proteases, prekallikrein (pKal) and factor XII (FXII), and a cofactor, high-molecular-weight kininogen (HK). Upon activation of the KKS, HK is cleaved to release bradykinin. Although the KKS is activated in humans and animals with inflammatory bowel disease (IBD), its role in the pathogenesis of IBD has not been characterized. In the present study, we determined the role of the KKS in the pathogenesis of IBD using mice that lack proteins involved in the KKS. In two colitis models, induced by dextran sulfate sodium (DSS) or 2,4,6-trinitrobenzene sulfonic acid (TNBS), mice deficient in HK, pKal, or bradykinin receptors displayed attenuated phenotypes, including body weight loss, disease activity index, colon length shortening, histological scoring, and colonic production of cytokines. Infiltration of neutrophils and inflammatory monocytes in the colonic lamina propria was reduced in HK-deficient mice. Reconstitution of HK-deficient mice through intravenous injection of HK recovered their susceptibility to DSS-induced colitis, increased IL-1ß levels in the colon tissue and bradykinin concentrations in plasma. In contrast to the phenotypes of other mice lacking other proteins involved in the KKS, mice lacking FXII had comparable colonic inflammation to that observed in wild-type mice. The concentration of bradykinin was significantly increased in the plasma of wild-type mice after DSS-induced colitis. In vitro analysis revealed that DSS-induced pKal activation, HK cleavage, and bradykinin plasma release were prevented by the absence of pKal or the inhibition of Kal. Unlike DSS, TNBS-induced colitis did not trigger HK cleavage. Collectively, our data strongly suggest that Kal, acting independently of FXII, contributes to experimental colitis by promoting bradykinin release from HK.


Assuntos
Bradicinina/metabolismo , Colite/imunologia , Colite/patologia , Sistema Calicreína-Cinina/imunologia , Cininogênio de Alto Peso Molecular/metabolismo , Pré-Calicreína/metabolismo , Animais , Bradicinina/sangue , Colite/induzido quimicamente , Sulfato de Dextrana , Fator XII/metabolismo , Interleucina-1beta/análise , Mucosa Intestinal/patologia , Cininogênio de Alto Peso Molecular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/genética , Neutrófilos/imunologia , Calicreína Plasmática/análise , Pré-Calicreína/genética , Receptores da Bradicinina/genética , Ácido Trinitrobenzenossulfônico
11.
Arterioscler Thromb Vasc Biol ; 26(10): 2260-6, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16902163

RESUMO

OBJECTIVE: Plasma high-molecular-weight kininogen (HK) is cleaved in inflammatory diseases by kallikrein to HKa with release of bradykinin (BK). We postulated a direct link between HKa and cytokine/chemokine release. METHODS AND RESULTS: HKa, but not BK, releases cytokines tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, and chemokines IL-8 and MCP-1 from isolated human mononuclear cells. At a concentration of 600 nM, glutathione-S-transferase (GST) fusion proteins of kininogen domain 3 (D3), a fragment of domain 3, E7P (aaG255-Q292), HK domain 5 (D5), the D5 recombinant peptides HG (aa K420-D474) and HGK (aa H475-S626) stimulated secretion of IL-1beta from mononuclear cells. Monoclonal antibodies (MAbs) specific for D5 or specific for D3 blocked release of IL-1beta by HKa, supporting the importance of both domains. Antibodies to HK receptors on leukocytes including Mac-1, LFA-1, uPAR, and C1qR inhibited IL-1beta secretion induced by tKa 98%, 89%, 85%, and 62%, respectively. Fractionation of mononuclear cells identified the responsible cell, a blood monocyte. Inhibitors of signaling pathways NFkB, JNK, and p38 but not extracellular signal-regulated kinase (ERK) decreased cytokine release from mononuclear cells. HKa increased the synthesis of IL-1beta as deduced by an increase of IL-1beta mRNA at 1 to 2 hours. CONCLUSIONS: HKa domains 3 and 5 may contribute to the pathogenesis of inflammatory diseases by releasing IL-1beta from human monocytes using intracellular signaling pathways initiated by uPAR, beta2 integrins and gC1qR.


Assuntos
Quimiocinas/metabolismo , Citocinas/metabolismo , Cininogênio de Alto Peso Molecular/farmacologia , Antígeno de Macrófago 1/metabolismo , Glicoproteínas de Membrana/metabolismo , Monócitos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Complemento/metabolismo , Anticorpos Monoclonais/farmacologia , Antígeno CD11a/imunologia , Humanos , Interleucina-1/antagonistas & inibidores , Interleucina-1/genética , Interleucina-1/metabolismo , Cininogênio de Alto Peso Molecular/imunologia , Cininogênio de Alto Peso Molecular/metabolismo , Antígeno de Macrófago 1/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , NF-kappa B/fisiologia , Concentração Osmolar , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/imunologia , Receptores de Complemento/imunologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Fatores de Tempo
12.
J Exp Med ; 214(9): 2649-2670, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28794132

RESUMO

In this study, we show that mice lacking high-molecular-weight kininogen (HK) were resistant to lipopolysaccharide (LPS)-induced mortality and had significantly reduced circulating LPS levels. Replenishment of HK-deficient mice with human HK recovered the LPS levels and rendered the mice susceptible to LPS-induced mortality. Binding of HK to LPS occurred through the O-polysaccharide/core oligosaccharide, consistent with the ability to bind LPS from K. pneumoniae, P. aeruginosa, S. minnesota, and different E. coli strains. Binding of LPS induced plasma HK cleavage to the two-chain form (HKa, containing a heavy chain [HC] and a light chain [LC]) and bradykinin. Both HKa and the LC, but not the HC, could disaggregate LPS. The light chain bound LPS with high affinity (K d = 1.52 × 10-9 M) through a binding site in domain 5 (DHG15). A monoclonal antibody against D5 significantly reduced LPS-induced mortality and circulating LPS levels in wild-type mice. Thus, HK, as a major LPS carrier in circulation, plays an essential role in endotoxemia.

13.
J Am Coll Cardiol ; 46(4): 707-13, 2005 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-16098440

RESUMO

OBJECTIVES: The purpose of this study was to test the hypothesis that activated monocytes with soluble plasma tissue factor (pTF) activate factors VII and X to generate thrombin. BACKGROUND: Despite heparin, thrombin is progressively generated during cardiac surgery with cardiopulmonary bypass (CPB), produces intravascular fibrin and fibrinolysis, and causes serious thromboembolic and nonsurgical bleeding complications. Thrombin is primarily produced in the surgical wound, but mechanisms are unclear. METHODS: In 13 patients, interactions of mononuclear cells, platelets, pTF, and pTF fractions to activate factors VII and X were evaluated in pre-bypass, perfusate, and pericardial wound blood before and during CPB. RESULTS: Monocytes are activated in wound, but not in pre-bypass or perfusate plasma (monocyte chemotactic protein-1 = 29.5 +/- 2.1 pmoles/l vs. 2.8 +/- 1.2 pmoles/l and 3.3 +/-1.4 pmoles/l, respectively). Wound pTF is substantially elevated compared to other locations (3.64 +/- 0.45 pmoles/l vs. 0.71 +/- 0.65 pmoles/l and 1.31 +/- 1.4 pmoles/l). Supernatant wound pTF contains 81.7% of TF antigen; wound microparticle pTF contains 18.3%. Wound monocytes and all C5a-stimulated monocytes (but not activated platelets) completely convert factor VII to factor VIIa with wound pTF. Activated monocytes more efficiently activate factor X with wound supernatant TF/factor VII(VIIa) complex than with wound microparticle TF/factor VII(fVIIa). The correlation coefficient (r) between wound thrombin generation (F1.2) and wound pTF concentration is 0.944 (p = 0.0004). CONCLUSIONS: During cardiac surgery with CPB, wound monocytes plus wound pTF or wound microparticle-free supernatant pTF preferentially accelerate activation of factor VII and factor X. This system represents a novel mechanism for thrombin generation via the TF coagulation pathway.


Assuntos
Coagulação Sanguínea/fisiologia , Ponte Cardiopulmonar/efeitos adversos , Fator VII/biossíntese , Fator X/biossíntese , Monócitos/fisiologia , Plasma/química , Hemorragia Pós-Operatória/fisiopatologia , Trombina/biossíntese , Tromboplastina/análise , Idoso , Idoso de 80 Anos ou mais , Fatores de Coagulação Sanguínea/fisiologia , Western Blotting , Fator VII/análise , Fator X/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Cuidados Pós-Operatórios , Cicatrização
14.
Curr Pharm Des ; 12(21): 2599-607, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16842160

RESUMO

High molecular weight kininogen (HK) is a plasma protein that is cleaved by plasma kallikrein in the clinical settings of sepsis and chronic inflammatory diseases such as rheumatoid arthritis and Crohn's disease. This proteolytic event results in a nonapeptide, bradykinin (BK), and a kinin-free derivative of HK, namely HKa. BK promotes angiogenesis by upregulation of bFGF through the B1 receptor or by stimulation of VEGF formation via the B2 receptor. Kininogen-deficient rats show diminished angiogenesis when neovascularization is stimulated. The formation of HKa results in exposure of domain 5 (D5). HKa or D5 inhibit endothelial cell migration and proliferation, both of which are needed for angiogenesis. In the chicken chorioallantoic membrane assay when neovascularization is stimulated by bFGF or VEGF, HKa or D5 inhibit angiogenesis. Monoclonal antibody C11C1, which prevents binding of HK to endothelial cells, also limits its conversion to BK thus downregulating angiogenesis. In vivo, mAb C11C1 inhibits tumor angiogenesis in mice as well as in experimental inflammatory arthritis and inflammatory bowel disease in Lewis rats. In vitro HKa or D5 inhibits endothelial cell adhesion to vitronectin and fibrinogen, resulting in anokis and apoptosis. The HKa receptor, uPAR, forms a signaling complex containing the integrin alphavbeta3 or alpha5beta1, caveolin, Src kinase Yes, focal adhesion kinase and paxcillin. HKa physically disrupts the complex by interfering with the binding of vitronectin to uPAR. Both mAb C11C1 and D5 have potential applications for controlling unwanted angiogenesis in inflammation and cancer.


Assuntos
Sistema Calicreína-Cinina/fisiologia , Neovascularização Patológica/fisiopatologia , Animais , Humanos , Inflamação/tratamento farmacológico , Inflamação/fisiopatologia , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Neovascularização Patológica/prevenção & controle
15.
Thromb Haemost ; 95(3): 462-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16525574

RESUMO

Soluble plasma tissue factor (TF) circulates in picomolar concentrations in healthy individuals and increases in a wide spectrum of diseases. This study tests the hypothesis that both truncated TF (rsTF) or soluble plasmaTF (pTF) in low concentration combine with monocytes or platelets to convert factorVII (fVII) to fVIIa. Both rsTF (33 kDa) and pTF (47 kDa), obtained from pericardial wounds of patients having cardiac surgery using cardiopulmonary bypass (CPB), were studied in association with blood cells and TF-bearing microparticles. Tissue factor was measured by ELISA. RsTF binds to erythrocytes, platelets, mononuclear cells and polymorphoneutrophils. The rate of fVII conversion with rsTF (1-10(3) nM) is highest with mononuclear cells, less with platelets, minimal with polymorphoneutrophils and undetectable with erythrocytes. Either stimulated or unstimulated mononuclear cells or platelets in the presence of 3.5 pM rsTF or pTF convert fVII (10 pM) [corrected] to fVIIa, but the amounts of fVIIa produced differ. When leukocytes or platelets are absent, microparticles associated with 3.5 pM TF antigen derived from pericardial wound plasma do not activate fVII. Stimulated mononuclear cells convert nearly all available fVII (10 nM) to fVIIa with 3.5 nM pTF; unstimulated mononuclear cells convert small amounts of fVII with 1 pM rsTF. In all comparisons mononuclear cells more efficiently convert fVII to fVIIa than do platelets. This study shows that stimulated mononuclear cells provide the most efficient platform for activation of rsTF or pTF at low concentrations of TF antigen.


Assuntos
Fator VIIa/metabolismo , Monócitos/efeitos dos fármacos , Tromboplastina/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Relação Dose-Resposta a Droga , Humanos , Cinética , Monócitos/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Tromboplastina/química , Tromboplastina/genética
16.
Circ Res ; 94(9): 1227-34, 2004 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-15044324

RESUMO

Cleaved high molecular weight kininogen (HKa) has been shown to inhibit in vivo neovascularization and induce apoptosis of endothelial cells. We have shown that HKa-induced apoptosis correlated with its antiadhesive effect and was regulated by extracellular matrix (ECM) proteins. In this study, we identified the urokinase-type plasminogen activator receptor (uPAR) as a target of HKa activity at the endothelial cell surface. Anti-uPAR antibodies blocked the apoptotic effect of HKa. Further studies revealed that uPAR formed a signaling complex containing integrin alpha(v)beta3 or alpha5beta1, caveolin, and Src kinase Yes in endothelial cells. HKa physically disrupted the formation of this complex in a manner that paralleled its apoptotic effect. For the first time, our results provide a mechanistic explanation for the previous observation that HKa selectively induces apoptosis of endothelial cells grown on vitronectin, but not cells grown on fibronectin. These data also resolve the controversial role of uPAR in mediating the apoptotic and antiadhesive activities of HKa.


Assuntos
Apoptose/efeitos dos fármacos , Cininogênio de Alto Peso Molecular/fisiologia , Receptores de Superfície Celular/fisiologia , Quinases da Família src , Anticorpos Monoclonais/farmacologia , Apoptose/fisiologia , Caveolina 1 , Caveolinas/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas , Células Endoteliais , Endotélio Vascular , Proteínas da Matriz Extracelular/fisiologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibronectinas/farmacologia , Humanos , Integrina alfa5beta1/fisiologia , Integrina alfaVbeta3/fisiologia , Cininogênio de Alto Peso Molecular/farmacologia , Substâncias Macromoleculares , Fosfatidilinositol Diacilglicerol-Liase/farmacologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-yes , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Veias Umbilicais , Vitronectina/farmacologia
17.
Thromb Haemost ; 94(3): 606-14, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16268479

RESUMO

Domain 5 (D5) of cleaved high molecular weight kininogen (HKa) inhibits angiogenesis in vivo and endothelial cell migration in vitro, but the cell signaling pathways involved in HKa and D5 inhibition of endothelial cell migration are incompletely delineated. This study examines the mechanism of HKa and D5 inhibition of two potent stimulators of endothelial cell migration, sphingosine 1-phosphate (S1P) and vascular endothelial growth factor (VEGF), that act through the P13-kinase-Akt signaling pathway. HKa and D5 inhibit bovine pulmonary artery endothelial cell (BPAE) or human umbilical vein endothelial cell chemotaxis in the modified-Boyden chamber in response toVEGF or S1P. The inhibition of migration by HKa is reversed by antibodies to urokinase-type plasminogen activator receptor. Both HKa and D5 decrease the speed of BPAE cell migration and alter the morphology in live, time-lapse microscopy after stimulation with S1P or VEGF. HKa and D5 reduce the localization of paxillin to the focal adhesions after S1P and VEGF stimulation. To better understand the intracellular signaling pathways, we examined the effect of HKa on the phosphorylation of Akt and its downstream effector, GSK-3alpha HKa and D5 inhibit phosphorylation of Akt and GSK-3alpha after stimulation withVEGF and S1P. Inhibitors of Akt and P13-kinase, the upstream activator of Akt, block endothelial cell migration and disrupt paxillin localization to the focal adhesions after stimulation with VEGF and S1P. Therefore we suggest that HKa through its D5 domain alters P13-kinase-Akt signaling to inhibit endothelial cell migration through alterations in the focal adhesions.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Cininogênio de Alto Peso Molecular/farmacologia , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Bovinos , Células Cultivadas , Cromonas/farmacologia , Células Endoteliais/metabolismo , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Cininogênio de Alto Peso Molecular/química , Lisofosfolipídeos/farmacologia , Morfolinas/farmacologia , Fragmentos de Peptídeos/química , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Estrutura Terciária de Proteína , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Wortmanina
18.
Arch Med Res ; 36(1): 87-95, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15900628

RESUMO

Inflammation is accompanied by activation of the plasma kallikrein-kinin system (KKS). KKS activation has been demonstrated in a variety of inflammatory human diseases. To further explore the participation of KKS in arthritis and inflammatory bowel disease, we used two experimental animal models in arthritis and enterocolitis. We found that activation of KKS is associated with arthritis induced by intraperitoneal injection of peptidoglycan-polysaccharide polymers (PG-PS) as well as the enterocolitis and systemic inflammation induced also by PG-PS when injected into the intestinal wall of genetically susceptible Lewis rats. We postulated that KKS participates in the pathogenesis of inflammatory reactions involved in cellular injury, coagulation, fibrinolysis, kinin formation, complement activation, cytokine secretion, and release of proteases. We demonstrated that therapy with a specific plasma kallikrein inhibitor modulated the experimental enterocolitis, arthritis, and systemic inflammation. The fact that deficiency of plasma high molecular weight kininogen in the genetically susceptible Lewis rat results in decreased chronic enterocolitis and systemic inflammation also supports our hypothesis. We suggest that KKS plays a similar role in idiopathic human intestinal inflammatory disease and arthritis, making kallikrein-kinin system proteins appealing targets for drug therapy in chronic inflammatory diseases such as rheumatoid arthritis and Crohn's disease.


Assuntos
Inflamação/sangue , Intestinos , Sistema Calicreína-Cinina/imunologia , Cininogênios/sangue , Cininogênios/química , Sequência de Aminoácidos , Animais , Artrite Experimental/metabolismo , Humanos , Intestinos/imunologia , Intestinos/patologia , Cininogênios/genética , Dados de Sequência Molecular , Peso Molecular , Estrutura Secundária de Proteína
19.
Curr Mol Med ; 3(4): 387-92, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12776993

RESUMO

Leukocyte recruitment to sites of inflammation, infection or vascular injury is a complex event, depending on a tightly coordinated sequence of leukocyte-endothelial- and leukocyte-platelet interactions, which are controlled by the expression and activation of various adhesion receptors and protease systems. The present review will focus on novel aspects of the regulation of integrin-dependent leukocyte adhesion by haemostatic factors and bacterial products. In particular, after a short overview of leukocyte recruitment, the review (i) will focus on the crosstalk between haemostatic factors and adhesion molecules with respect to leukocyte extravasation based on the paradigms of the urokinase receptor and high molecular weight kininogen, (ii) will provide information on novel mechanisms for the regulation of leukocyte recruitment by bacterial proteins, on the basis of the anti-inflammatory role of Staphylococcus aureus extracellular adhesive protein and (iii) will draw attention to the junctional adhesion molecules, a novel family of adhesive receptors that are counter-receptors for leukocyte integrins and mediate vascular cell interactions. The better understanding of the interactions between vascular cells and particularly of integrin-dependent leukocyte adhesion may lead to the development of novel therapeutical concepts in inflammatory vascular disorders.


Assuntos
Bactérias/metabolismo , Inflamação/imunologia , Integrinas/metabolismo , Leucócitos/citologia , Animais , Adesão Celular , Endotélio Vascular/metabolismo , Hemostasia , Humanos , Cininogênios/metabolismo , Modelos Biológicos , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Staphylococcus aureus/metabolismo
20.
Mol Immunol ; 41(8): 759-66, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15234555

RESUMO

A growing body of evidence supports the hypothesis that atherosclerosis has an inflammatory component, and that immune mechanisms, including complement activation, are likely to be involved. gC1q-R/p33 (gC1q-R) is a multifunctional and multicompartmental cellular protein, which is postulated to play a role in inflammation and thrombosis by interacting with C1q and high molecular weight kininogen (HK). To examine the expression of gC1q-R and its major ligands, C1q and HK, in human atherosclerotic lesions, sections of carotid arteries removed during endarterectomy and coronary arteries obtained at autopsy were stained with specific polyclonal or monoclonal antibodies. Control sections were stained with irrelevant rabbit IgG or isotype matched murine monoclonal antibody (MOPC), respectively. Tissue sections were counterstained with hematoxylin and examined by light microscopy. Specific staining for gC1q-R, C1q, and HK was observed in and around atherosclerotic lesions. In contrast to control antibodies, antibodies directed against gC1q-R reacted with endothelial cells, foam cells, smooth muscle cells, and inflammatory cells present in the intima and media of atherosclerotic lesions. In addition, the necrotic central core of advanced lesions with calcifications, fibrin, and lipids, stained intensely for gC1q-R, and negligibly with control antibodies. HK demonstrated a similar staining pattern, whereas C1q was most heavily expressed in the fibrous cap and necrotic core of atherosclerotic lesions. The localization of gC1q-R and its ligands C1q and HK in atherosclerotic lesions, and the previously described ability of gC1q-R to modulate complement, kinin, and coagulation cascades, suggest that gC1q-R may play an important role in promoting inflammation and thrombosis in atherosclerotic lesions.


Assuntos
Arteriosclerose/metabolismo , Glicoproteínas de Membrana/genética , Receptores de Complemento/genética , Arteriosclerose/genética , Arteriosclerose/patologia , Artérias Carótidas/patologia , Humanos , Imuno-Histoquímica , Ligantes , Glicoproteínas de Membrana/biossíntese , Receptores de Complemento/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA