Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Alzheimers Dement ; 18(10): 1919-1929, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34978145

RESUMO

Increased activation of the contact system protein high molecular weight kininogen (HK) has been shown in plasma and cerebrospinal fluid of Alzheimer's disease (AD) patients, but its potential role in the brain has not been explored. We assessed HK levels in brain tissue from 20 AD patients and controls and modeled the effects of HK on microglia-like cells in culture. We show increased levels of HK in the hippocampus of AD patients, which colocalized with amyloid beta (Aß) deposits and activated microglia. Treatment of microglia with HK led to cell clustering and elevated levels of phagocytosed Aß. We demonstrate that microglia internalize HK and traffic it to lysosomes, which is accompanied by reduced activity of lysosomal cathepsins L and S. Our results suggest that HK accumulation in the AD hippocampus may alter microglial uptake and degradation of Aß fibrils, possibly contributing to microglial dysfunction in AD.


Assuntos
Doença de Alzheimer , Microglia , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Catepsinas/metabolismo , Catepsinas/farmacologia , Cininogênio de Alto Peso Molecular/metabolismo , Cininogênio de Alto Peso Molecular/farmacologia , Lisossomos/metabolismo , Microglia/metabolismo , Fagocitose
2.
J Biol Chem ; 286(28): 24561-71, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21586566

RESUMO

The nonenzymatic cofactor high molecular weight kininogen (HK) is a precursor of bradykinin (BK). The production of BK from HK by plasma kallikrein has been implicated in the pathogenesis of inflammation and vascular injury. However, the functional role of HK in the absence of prekallikrein (PK), the proenzyme of plasma kallikrein, on vascular endothelial cells is not fully defined. In addition, no clinical abnormality is seen in PK-deficient patients. Therefore, an investigation into the effect of HK, in the absence of PK, on human pulmonary artery endothelial cell (HPAEC) function was performed. HK caused a marked and dose-dependent increase in the intracellular calcium [Ca(2+)](i) level in HPAEC. Gd(3+) and verapamil potentiated the HK-induced increase in [Ca(2+)](i). HK-induced Ca(2+) increase stimulated endothelial nitric oxide (NO) and prostacyclin (PGI(2)) production. The inhibitors of B(2) receptor-dependent signaling pathway impaired HK-mediated signal transduction in HPAEC. HK had no effect on endothelial permeability at physiological concentration. This study demonstrated that HK regulates endothelial cell function. HK could play an important role in maintaining normal endothelial function and blood flow and serve as a cardioprotective peptide.


Assuntos
Cardiotônicos/farmacologia , Células Endoteliais/metabolismo , Cininogênio de Alto Peso Molecular/farmacologia , Receptor B2 da Bradicinina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Cálcio/metabolismo , Cardiotônicos/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Epoprostenol/biossíntese , Gadolínio/farmacologia , Humanos , Cininogênio de Alto Peso Molecular/metabolismo , Óxido Nítrico/biossíntese , Pré-Calicreína/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Transdução de Sinais/fisiologia , Vasodilatadores/farmacologia , Verapamil/farmacologia
3.
Biochem Biophys Res Commun ; 427(3): 497-502, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-23000411

RESUMO

High molecular weight kininogen (HK) is a plasma glycoprotein with multiple functions, including the regulation of coagulation. We previously demonstrated that domain 5 (D5(H)), a functional domain of HK, and its derived peptides played an important role in the vitronectin-mediated suppression of cancer cell adhesion and invasion. However, the underlying mechanisms of the D5(H)-mediated suppressive effects remain to be elucidated. Here, we showed that D5(H) and its derivatives inhibited the collagen-mediated cell adhesion and invasion of human osteosarcoma MG63 cells. Using purified D5(H) fused to glutathione-S-transferase (GST) and D5(H)-derived peptides for column chromatography, an actin-binding protein, α-actinin-4, was identified as a binding protein of D5(H) with high-affinity for P-5m, a core octapeptide of D5(H). Immunofluorescence microscopy demonstrated that D5(H) co-localized with α-actinin-4 inside MG63 cells. In addition, exogenous GST-D5(H) added to the culture media was transported into MG63 cells, although GST alone as a control was not. As α-actinin-4 regulates actin polymerization necessary for cell adhesion and is related to the integrin-dependent attachment of cells to the extracellular matrix, our results suggest that D5(H) may modulate cell adhesion and invasion together with actinin-4.


Assuntos
Actinina/metabolismo , Cininogênio de Alto Peso Molecular/metabolismo , Neoplasias/patologia , Sequência de Aminoácidos , Adesão Celular , Linhagem Celular Tumoral , Colágeno/metabolismo , Humanos , Cininogênio de Alto Peso Molecular/genética , Cininogênio de Alto Peso Molecular/farmacologia , Dados de Sequência Molecular , Invasividade Neoplásica , Neoplasias/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia
4.
Proc Natl Acad Sci U S A ; 106(2): 570-5, 2009 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-19126685

RESUMO

Angiogenesis, the synthesis of new blood vessels from preexisting vessels, plays a critical role in normal wound healing and tumor growth. HKa (cleaved high molecular weight kininogen) is an endogenous inhibitor of angiogenesis formed by the cleavage of kininogen on endothelial cells. Ferritin is a protein principally known for its central role in iron storage. Here, we demonstrate that ferritin binds to HKa with high affinity (K(d) 13 nM). Further, ferritin antagonizes the antiangiogenic effects of HKa, enhancing the migration, assembly, and survival of HKa-treated endothelial cells. Effects of ferritin were independent of its iron content. Peptide mapping revealed that ferritin binds to a 22-aa subdomain of HKa that is critical to its antiangiogenic activity. In vivo, ferritin opposed HKa's antiangiogenic effects in a human prostate cancer xenograft, restoring tumor-dependent vessel growth. Ferritin-mediated regulation of angiogenesis represents a new angiogenic regulatory pathway, and identifies a new role for ferritin in cell biology.


Assuntos
Ferritinas/metabolismo , Cininogênio de Alto Peso Molecular/metabolismo , Neovascularização Patológica , Movimento Celular , Células Endoteliais/fisiologia , Ferritinas/farmacologia , Humanos , Cininogênio de Alto Peso Molecular/farmacologia , Masculino , Neoplasias da Próstata/patologia , Ligação Proteica , Mapeamento de Interação de Proteínas
5.
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
6.
Arterioscler Thromb Vasc Biol ; 29(10): 1602-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19661487

RESUMO

OBJECTIVE: Factor XI (FXI) promotes hemostasis and thrombosis through enhancement of thrombin generation and has been shown to play a critical role in the formation of occlusive thrombi in arterial injury models. The aim of this study was to investigate the mechanisms governing interactions between FXI and platelets. METHODS AND RESULTS: Platelet adhesion to immobilized FXI was abrogated in the presence of the low-density lipoprotein (LDL) receptor antagonist, receptor-associated protein (RAP), soluble recombinant apolipoprotein E receptor 2 (ApoER2), or the LDL-binding domain 1 or 2 of ApoER2. FXI supported wild-type murine platelet binding; in contrast, ApoER2-deficient murine platelets did not adhere to FXI. In the presence of shear, platelet aggregates formed on FXI or activated FXI (FXIa) surfaces, whereas the presence of RAP, binding domain 1 of ApoER2, or an anti-GPIb alpha mAb blocked platelet adhesion to FXI or FXIa under shear. Soluble FXI bound to immobilized ApoER2' with an affinity of 61 nmol/L. CONCLUSIONS: This study has identified apolipoprotein E receptor 2 (ApoER2, LRP8), a member of the LDL receptor family, as a platelet receptor for FXI. The interaction of FXI with other cell types that express ApoER2 remains to be explored.


Assuntos
Fator XI/metabolismo , Receptores de Lipoproteínas/metabolismo , Animais , Plaquetas/metabolismo , Cálcio/metabolismo , Humanos , Cininogênio de Alto Peso Molecular/farmacologia , Proteínas Relacionadas a Receptor de LDL , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Adesividade Plaquetária , Zinco/farmacologia
7.
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
8.
J Biochem ; 140(6): 825-30, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17071946

RESUMO

High molecular weight (HMW) kininogen is known to be a large plasma protein and cleaved by plasma proteinase kallikrein, then it generates four fragments in the blood coagulation cascade: heavy chain, bradykinin, fragment 1.2, and light chain. The fragment 1.2 has also been found in the basic protein fraction of bovine milk as a bioactive protein which promotes osteoblast proliferation. The milk basic protein has been shown to be a multi functional edible protein which promotes bone formation and inhibits bone resorption. In the present study, we purified the fragment 1.2 from bovine plasma and assessed it could promote osteoblast proliferation and posses the activity after pepsin digestion. Purified plasma HMW kininogen did not promote the proliferation, however, the kallikrein-cleaved HMW kininogen promoted the proliferation. The fragment 1.2, purified from the proteolysate, also promoted the proliferation. The pepsin digestion was performed according to the method of the assessment of allergenesity of genetically modified crops. After pepsin digestion, the fragment 1.2 generated resistant fragments and showed the promoting activity of osteoblast proliferation. These results suggest that the enzymatically-digested fragments of bovine HMW kininogen are able to be a naturally occurred active protein that promotes the bone formation by oral administration.


Assuntos
Cininogênio de Alto Peso Molecular/farmacologia , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Células 3T3 , Animais , Bradicinina/farmacologia , Bovinos , Proliferação de Células/efeitos dos fármacos , Calicreínas/metabolismo , Camundongos , Osteogênese/efeitos dos fármacos
9.
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
10.
Curr Cancer Drug Targets ; 5(7): 519-28, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16305348

RESUMO

High molecular weight kininogen (HK) is an abundant, multi-domain plasma protein that circulates in plasma primarily in its single chain form. Proteolytic cleavage of HK by plasma kallikrein releases the vasoactive nanopeptide bradykinin (BK), and converts HK into two-chain HK (HKa). BK appears to have pro-angiogenic activity, most likely mediated through binding to B1 and B2 receptors on endothelial cells. Conversely, HKa and its domain 5, but not (single chain) HK, have potent anti-angiogenic activity comparable to other endogenous angiogenesis inhibitors. The mechanism by which HKa exerts its anti-angiogenic activity remains controversial, but appears to involve binding to cell surface tropomyosin and induction of apoptosis of proliferating endothelial cells. A role for tropomyosin in mediating the anti-angiogenic signals of other anti-angiogenic proteins such as endostatin and histidine-proline-rich glycoprotein (HPRG) has also been reported. Here we review the physiological importance of high molecular weight kininogen in angiogenesis, with emphasis on the mechanism(s) by which this activity is mediated.


Assuntos
Cininogênio de Alto Peso Molecular/farmacologia , Neovascularização Patológica/prevenção & controle , Fragmentos de Peptídeos/farmacologia , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacologia , Humanos , Cininogênio de Alto Peso Molecular/química , Cininogênio de Alto Peso Molecular/metabolismo , Dados de Sequência Molecular , Calicreína Plasmática/metabolismo
11.
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
12.
FASEB J ; 15(13): 2365-76, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11689462

RESUMO

Proteolytic cleavage of single-chain, high molecular weight kininogen (HK) by kallikrein releases the short-lived vasodilator bradykinin and leaves behind a two-chain, high molecular weight kininogen (HKa) reported to bind to the beta2-integrin Mac-1 (CR3, CD11b/CD18, alphaMbeta2) on neutrophils and exert antiadhesive properties by binding to the urokinase receptor (uPAR) and vitronectin. We define the molecular mechanisms for the antiadhesive effects of HK related to disruption of beta2-integrin-mediated cellular interactions in vitro and in vivo. In a purified system, HK and HKa inhibited the binding of soluble fibrinogen and ICAM-1 to immobilized Mac-1, but not the binding of ICAM-1 to immobilized LFA-1 (CD11a/CD18, alphaLbeta2). This inhibitory effect could be attributed to HK domain 5 and to a lesser degree to HK domain 3, consistent with the requirement of both domains for binding to Mac-1. Accordingly, HK, HKa, and domain 5 inhibited the adhesion of Mac-1 but not LFA-1-transfected K562 human erythroleukemic cells to ICAM-1. Moreover, adhesion of human monocytic cells to fibrinogen and to human endothelial cells was blocked by HK, HKa, and domain 5. By using peptides derived from HK domain 5, the sequences including amino acids H475-G497 (and to a lesser extent, G440-H455) were identified as responsible for the antiadhesive effect, which was independent of uPAR. Finally, administration of domain 5 into mice, followed by induction of thioglycollate-provoked peritonitis, decreased the recruitment of neutrophils by approximately 70% in this model of acute inflammation. Taken together, HKa (and particularly domain 5) specifically interacts with Mac-1 but not with LFA-1, thereby blocking Mac-1-dependent leukocyte adhesion to fibrinogen and endothelial cells in vitro and in vivo and serving as a novel endogenous regulator of leukocyte recruitment into the inflamed tissue.


Assuntos
Cininogênio de Alto Peso Molecular/farmacologia , Leucócitos/efeitos dos fármacos , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Ligação Competitiva/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Fibrinogênio/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Células K562 , Cininogênio de Alto Peso Molecular/química , Leucócitos/citologia , Leucócitos/metabolismo , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Infiltração de Neutrófilos/efeitos dos fármacos , Peritonite/induzido quimicamente , Peritonite/patologia , Peritonite/prevenção & controle , Plasmídeos/genética , Tioglicolatos/administração & dosagem , Células U937
13.
Arterioscler Thromb Vasc Biol ; 21(9): 1427-33, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11557667

RESUMO

We recently reported that domain 5 (D5) of high-molecular-weight kininogen inhibited critical steps required for angiogenesis. Thus, it was named kininostatin. To understand its mechanism of action, we further investigated the effects of D5 on basic fibroblast growth factor (bFGF)-induced endothelial cell proliferation and cell viability. We report here that D5-inhibited cell proliferation of human endothelial cells stimulated by bFGF was associated with a significant reduction of cyclin D1 expression, which is a critical component required for the transition from G(1) to S phase of the cell cycle. However, inhibition of cell proliferation by D5 was not due to an inhibition of extracellular signal-regulated protein kinase activity. Endothelial cells underwent apoptosis when cultured in a serum-free medium, which was prevented by bFGF. D5 reversed the protective effect of bFGF by 80%. Cells treated with D5 in the presence of bFGF showed typical morphological features of apoptosis, which was further confirmed by 2 additional assays: Hoechst 33258 cell staining and DNA fragmentation analysis. We conclude that the inhibition of endothelial cell proliferation and induction of apoptosis together represent a major contribution to the antiangiogenic activity of D5.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose , Endotélio Vascular/citologia , Cininogênio de Alto Peso Molecular/química , Cininogênio de Alto Peso Molecular/farmacologia , Neovascularização Fisiológica , Inibidores da Angiogênese/química , Divisão Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Ciclina D1/metabolismo , DNA/biossíntese , Fragmentação do DNA , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estrutura Terciária de Proteína
14.
Biomaterials ; 37: 1-12, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25443791

RESUMO

High-resolution electrophoresis of FXII-derived proteins produced by contact activation of FXII in buffer solutions (i.e. in absence of plasma proteins) with hydrophilic and silanized-glass activators spanning the observable range of water wettability (hydrophilic to hydrophobic), shows no evidence of proteolytic cleavage of FXII into αFXIIa or ßFXIIa. The autoactivation mixture contains only a single-chain protein with a molecular weight of ∼80 kDa, confirming Oscar Ratnoff's previous finding of a single-chain activated form of FXII that he called 'HFea'. Functional assays have shown that these autoactivation products exhibit procoagulant potential (protease activity inducing clotting of blood) or amidolytic potential (cleaves amino bonds in s-2302 chromogen but do not cause coagulation of plasma) or both amidolytic potential and procoagulant potential. Some of these proteins also have the remarkable potential to 'suppress autoactivation' (i.e. suppress creation of enzymes with procoagulant potential). It is thus hypothesized that autoactivation of FXII in the absence of plasma proteins generates not just a single type of activated conformer, as suggested by previous researchers, but rather an ensemble of conformer products with collective activity that varies with activator surface energy used in contact activation of FXII. Furthermore, reaction of αFXIIa with FXII in buffer solution does not produce additional αFXIIa by the putative autoamplification reaction FXIIa + FXII â†’ 2FXIIa as has been proposed in past literature to account for the discrepancy between chromogenic and plasma-coagulation assays for αFXIIa in buffer solution. Instead, net procoagulant activity measured directly by plasma-coagulation assays, decreases systematically with increasing FXII solution concentration. Under the same reaction conditions, chromogenic assay reveals that net amidolytic activity increases with increasing FXII solution concentration. Thus, although autoamplification does not occur it appears that there is some form of "FXII self reaction" that influences products of αFXIIa reaction with FXII. Electrophoretic measurements indicate that no proteolytic cleavage takes in this reaction leading us to conclude that change in activity is most likely due to change(s) in FXII conformation (with related change in enzyme activity).


Assuntos
Materiais Biocompatíveis/farmacologia , Fator XII/química , Fator XII/metabolismo , Hematologia , Coagulação Sanguínea/efeitos dos fármacos , Soluções Tampão , Eletroforese , Ativação Enzimática/efeitos dos fármacos , Fator XIIa/metabolismo , Humanos , Hidrólise/efeitos dos fármacos , Cininogênio de Alto Peso Molecular/farmacologia , Pré-Calicreína/farmacologia , Pesquisa , Soluções , Fatores de Tempo
15.
Thromb Haemost ; 114(3): 603-13, 2015 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-26063414

RESUMO

Cleaved high-molecular-weight kininogen (HKa) or its peptide domain 5 (D5) alone exert anti-adhesive properties in vitro related to impeding integrin-mediated cellular interactions. However, the anti-adhesive effects of HKa in vivo remain elusive. In this study, we investigated the effects of HKa on leukocyte recruitment and neointima formation following wire-induced injury of the femoral artery in C57BL/6 mice. Local application of HKa significantly reduced the accumulation of monocytes and also reduced neointimal lesion size 14 days after injury. Moreover, C57BL/6 mice transplanted with bone marrow from transgenic mice expressing enhanced green fluorescence protein (eGFP) showed a significantly reduced accumulation of eGFP+-cells at the arterial injury site and decreased neointimal lesion size after local application of HKa or the polypeptide D5 alone. A differentiation of accumulating eGFP+-cells into highly specific smooth muscle cells (SMC) was not detected in any group. In contrast, application of HKa significantly reduced the proliferation of locally derived neointimal cells. In vitro, HKa and D5 potently inhibited the adhesion of SMC to vitronectin, thus impairing their proliferation, migration, and survival rates. In conclusion, application of HKa or D5 decreases the inflammatory response to vascular injury and exerts direct effects on SMC by impeding the binding of integrins to extracellular matrix components. Therefore, HKa and D5 may hold promise as novel therapeutic substances to prevent neointima formation.


Assuntos
Cininogênio de Alto Peso Molecular/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima , Fragmentos de Peptídeos/farmacologia , Lesões do Sistema Vascular/prevenção & controle , Animais , Transplante de Medula Óssea , Proliferação de Células/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Artéria Femoral/efeitos dos fármacos , Artéria Femoral/lesões , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Integrinas/metabolismo , Cininogênio de Alto Peso Molecular/genética , Cininogênio de Alto Peso Molecular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/lesões , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Fatores de Tempo , Células U937 , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia , Vitronectina/metabolismo
16.
J Thromb Haemost ; 1(1): 164-70, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12871554

RESUMO

Previously we demonstrated that domain 5 (D5) of high-molecular-weight kininogen (HK) inhibits neovascularization in the chicken chorioallantoic membrane (CAM) assay and further found that kallikrein cleaved HK (HKa) inhibited FGF2-and VEGF-induced neovascularization, and thus was antiangiogenic. In this study, we sought to demonstrate whether uncleaved HK stimulates neovascularization and thus is proangiogenic. The chick chorioallantoic membrane was used as an in ovo assay of angiogenesis. Low-molecular-weight kininogen stimulates angiogenesis, indicating that D5 is not involved. Bradykinin stimulates neovascularization equally to HK and LK and is likely to be responsible for the effect of HK. A murine monoclonal antibody to HK (C11C1) also recognizes a similar component in chicken plasma as detected by surface plasmon resonance. Angiogenesis induced by FGF2 and VEGF is inhibited by this monoclonal antibody and is a more potent inhibitor of neovascularization induced by VEGF than an integrin alphavbeta3 antibody (LM 609). Our postulate that C11C1 inhibits the stimulation of angiogenesis by HK was confirmed when either C11C1 or D5 completely inhibited angiogenesis in the CAM induced by HK. Growth of human fibrosarcoma (HT-1080) on the CAM was inhibited by GST-D5 and C11C1. These results indicate HK is proangiogenic probably by releasing bradykinin and that a monoclonal antibody directed to HK could serve as an antiangiogenic agent with a potential for inhibiting tumor angiogenesis and other angiogenesis-mediated disorders.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Cininogênio de Alto Peso Molecular/antagonistas & inibidores , Neovascularização Fisiológica/imunologia , Alantoide/irrigação sanguínea , Animais , Anticorpos Monoclonais/imunologia , Bradicinina/farmacologia , Embrião de Galinha , Córion/irrigação sanguínea , Relação Dose-Resposta a Droga , Fatores de Crescimento Endotelial/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibrossarcoma/metabolismo , Glutationa Transferase/genética , Glutationa Transferase/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Cininogênio de Alto Peso Molecular/imunologia , Cininogênio de Alto Peso Molecular/farmacologia , Cininogênio de Baixo Peso Molecular/farmacologia , Linfocinas/farmacologia , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Ressonância de Plasmônio de Superfície , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
17.
Thromb Haemost ; 92(4): 811-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15467913

RESUMO

Evidence is accumulating to suggest that TFPI-2 is involved in regulating pericellular proteases implicated in a variety of physiologic and pathologic processes including cancer cell invasion, vascular inflammation, and atherosclerosis. Recent immunohistochemical studies of advanced atherosclerotic lesions, demonstrated a similar tissue distribution for TFPI-2, High Molecular Weight Kininogen (HK), and gC1qR/p33 (gC1qR), a ubiquitously expressed, multicompartmental cellular protein involved in modulating complement, coagulation, and kinin cascades. Further studies to evaluate TFPI-2 interactions with gC1qR demonstrated direct interactions between gC1qR and TFPI-2 using immunoprecipitation and solid phase binding studies. Specific and saturable binding between TFPI-2 and gC1qR (estimated Kd: approximately 70 nM) was observed by ELISA and surface plasmon resonance (Biacore) binding assays. Binding was inhibited by antibodies to gC1qR, and was strongly dependent on the Kunitz-2 domain of TFPI-2, as deletion of this domain reduced gC1qR-TFPI-2 interactions by approximately 75%. Deletion of gC1qR amino acids 74-95, involved in C1q binding, had no effect on gC1qR binding to TFPI-2, although antibodies to this region and purified C1q both inhibited binding, most likely via allosteric effects. In contrast, HK did not affect TFPI-2 binding to gC1qR. Binding of TFPI-2 to gC1qR produced statistically significant but modest reductions in TFPI-2 inhibition of plasmin, but had no effect on kallikrein inhibition in fluid phase chromogenic assays. Taken together, these data suggest that gC1qR may participate in tissue remodeling and inflammation by localizing TFPI-2 to the pericellular environment to modulate local protease activity and regulate HK activation.


Assuntos
Endotélio Vascular/patologia , Glicoproteínas/metabolismo , Receptores de Hialuronatos/metabolismo , Inflamação/etiologia , Proteínas de Transporte , Complemento C1q/farmacologia , Fibrinolisina/antagonistas & inibidores , Glicoproteínas/farmacologia , Humanos , Receptores de Hialuronatos/farmacologia , Calicreínas/antagonistas & inibidores , Cininogênio de Alto Peso Molecular/farmacologia , Proteínas Mitocondriais , Ligação Proteica/efeitos dos fármacos
18.
Thromb Haemost ; 85(3): 544-51, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11307829

RESUMO

Biotin-FXI optimally bound to HUVEC in the presence of 40 nM high molecular weight kininogen (HK) and > or =7 microM Zn2+. There was little specific FXI binding in the absence of added HK and at concentrations of Zn2+ <15 microM. FXI and prekallikrein, but not prothrombin, blocked biotin-FXI binding to HUVEC in the presence of HK with an IC50 of 18 nM and 180 nM, respectively. Monoclonal antibody HKL16 and peptide SDD31 also inhibited biotin-XI binding in the presence of HK with an IC50 of 4.7 nM and 50 microM, respectively. Alternatively, peptide T249-F260 of FXI's apple domain 3 and heparin monosulfate were weak inhibitors of FXI binding to HUVEC. FXI bound to HUVEC with an apparent Kd of 6.9 +/- 3.0 nM and Bmax of 13 +/- 2.6 x 10(6) sites/cell. FXI bound to HK on HUVEC, but not prothrombin, became converted to FXIa. FXI activation on HUVEC resulted from tissue culture media bovine factor XIIa. HUVEC grown in human factor XI-deficient serum did not support FXI activation. FXI binding to HUVEC in culture was mostly mediated by HK and FXI activation on HUVEC is dependent on cell-associated factor XIIa.


Assuntos
Endotélio Vascular/citologia , Fator XI/metabolismo , Ligação Competitiva , Biotina/metabolismo , Técnicas de Cultura de Células , Meios de Cultura/farmacologia , Endotélio Vascular/metabolismo , Fator XI/biossíntese , Fator XII/farmacologia , Humanos , Cinética , Cininogênio de Alto Peso Molecular/farmacologia , Ligação Proteica/efeitos dos fármacos , Protrombina/farmacologia , Veias Umbilicais
19.
Thromb Haemost ; 85(1): 119-24, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11204562

RESUMO

Although proteins of the kinin-forming pathway are bound along the surface of endothelial cells, the mechanism of activation of this proteolytic cascade is unclear. Endothelial cell surface proteins, gC1qR and cytokeratin 1, are capable of binding Factor XII and high molecular weight kininogen (HK) in a zinc-dependent reaction thus we considered the possibility that these proteins might catalyze initiation of the cascade. Incubation of Factor XII, prekallikrein, and HK with gC1qR or cytokeratin 1 leads to a zinc-dependent and Factor XII-dependent conversion of prekallikrein to kallikrein. We also demonstrate that normal plasma is capable of activating upon interaction with the cells whereas plasma deficient in Factor XII, prekallikrein and HK do not activate. Normal plasma activation was inhibitable by antibody to gC1qR and cytokeratin 1. Thus, gC1qR and cytokeratin 1, represent potential initiating surfaces for activation of the plasma kinin-forming cascade and may do so as a result of their expression along cell surfaces.


Assuntos
Endotélio Vascular/metabolismo , Fator XII/metabolismo , Fator XII/farmacologia , Receptores de Hialuronatos , Sistema Calicreína-Cinina/efeitos dos fármacos , Queratinas/farmacologia , Glicoproteínas de Membrana , Receptores de Complemento/metabolismo , Proteínas de Transporte , Proteínas Inativadoras do Complemento 1/farmacologia , Relação Dose-Resposta a Droga , Endotélio Vascular/química , Endotélio Vascular/citologia , Fator XII/efeitos dos fármacos , Humanos , Queratinas/metabolismo , Cinética , Cininogênio de Alto Peso Molecular/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/farmacologia , Pré-Calicreína/metabolismo , Pré-Calicreína/farmacologia , Veias Umbilicais/citologia , Zinco/farmacologia
20.
Biomaterials ; 21(4): 405-14, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10656323

RESUMO

Adsorbed proteins on biomaterial surfaces determine whether cells adhere, but rheological variables are also critical. Neutrophil adhesion under well-defined radial flow conditions was studied on glass preadsorbed with plasma proteins or plasma protein domain fragments. Fibrinogen, low-molecular-weight kininogen (LK), high-molecular-weight kininogen (HK), cleaved HK (HKa), and recombinant HK domains 3 and 5 (D3 and D5H) were used. The number of adherent cells on the HK and HKa surfaces was less than 10% that found on the fibrinogen absorbed surface. The degree of spreading was minimal and detachment of adherent neutrophils was observed. HK and HKa contain binding sites for both anionic surfaces and neutrophils in the same domain (D5H). When adsorbed to surfaces, HK and HKa did not have the neutrophil binding sites available and therefore exhibited an anti-adhesive effect. Although D5H contains anionic surface binding sites, its small molecular size required a higher number of adsorbed molecules to cover the surface before a significant decrease in cell adhesion was observed. Since LK and D3 do not possess specific anionic surface binding sites, the adsorption of these proteins on glass was very low compared to HK and HKa. Thus, extensive cell adhesion and spreading were observed on the surfaces partially covered with preadsorbed LK and D3.


Assuntos
Materiais Revestidos Biocompatíveis/farmacologia , Vidro , Cininogênio de Alto Peso Molecular/farmacologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Adsorção , Adesão Celular , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/metabolismo , Fibrinogênio/química , Fibrinogênio/metabolismo , Fibrinogênio/farmacologia , Humanos , Cininogênio de Alto Peso Molecular/sangue , Cininogênio de Alto Peso Molecular/química , Cininogênio de Baixo Peso Molecular/sangue , Cininogênio de Baixo Peso Molecular/química , Cininogênio de Baixo Peso Molecular/farmacologia , Ativação de Neutrófilo , Neutrófilos/metabolismo , Ligação Proteica , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Reologia , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA