Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros












Intervalo de año de publicación
1.
Blood Adv ; 7(7): 1156-1167, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36409609

RESUMEN

A dysregulated plasma contact system is involved in various pathological conditions, such as hereditary angioedema, Alzheimer disease, and sepsis. We previously showed that the 3E8 anti-high molecular weight kininogen (anti-HK) antibody blocks HK cleavage and bradykinin generation in human plasma ex vivo. Here, we show that 3E8 prevented not only HK cleavage but also factor XI (FXI) and prekallikrein (PK) activation by blocking their binding to HK in mouse plasma in vivo. 3E8 also inhibited contact system-induced bradykinin generation in vivo. Interestingly, FXII activation was also inhibited, likely because of the ability of 3E8 to block the positive feedback activation of FXII by kallikrein (PKa). In human plasma, 3E8 also blocked PK and FXI binding to HK and inhibited both thrombotic (FXI activation) and inflammatory pathways (PK activation and HK cleavage) of the plasma contact system activation ex vivo. Moreover, 3E8 blocked PKa binding to HK and dose-dependently inhibited PKa cleavage of HK. Our results reveal a novel strategy to inhibit contact system activation in vivo, which may provide an effective method to treat human diseases involving contact system dysregulation.


Asunto(s)
Precalicreína , Trombosis , Humanos , Animales , Ratones , Precalicreína/química , Precalicreína/metabolismo , Factor XI/metabolismo , Bradiquinina/farmacología , Bradiquinina/química , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo
3.
J Thromb Haemost ; 17(12): 2131-2140, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31420909

RESUMEN

BACKGROUND: Inorganic polyphosphate modulates the contact pathway of blood clotting, which is implicated in thrombosis and inflammation. Polyphosphate polymer lengths are highly variable, with shorter polymers (approximately 60-100 phosphates) secreted from human platelets, and longer polymers (up to thousands of phosphates) in microbes. We previously reported that optimal triggering of clotting via the contact pathway requires very long polyphosphates, although the impact of shorter polyphosphate polymers on individual proteolytic reactions of the contact pathway was not interrogated. OBJECTIVES AND METHODS: We conducted in vitro measurements of enzyme kinetics to investigate the ability of varying polyphosphate sizes, together with high molecular weight kininogen and Zn2+ , to mediate four individual proteolytic reactions of the contact pathway: factor XII autoactivation, factor XII activation by kallikrein, prekallikrein activation by factor XIIa, and prekallikrein autoactivation. RESULTS: The individual contact pathway reactions were differentially dependent on polyphosphate length. Very long-chain polyphosphate was required to support factor XII autoactivation, whereas platelet-size polyphosphate significantly accelerated the activation of factor XII by kallikrein, and the activation of prekallikrein by factor XIIa. Intriguingly, polyphosphate did not support prekallikrein autoactivation. We also report that high molecular weight kininogen was required only when kallikrein was the enzyme (ie, FXII activation by kallikrein), whereas Zn2+ was required only when FXII was the substrate (ie, FXII activation by either kallikrein or FXIIa). Activation of prekallikrein by FXIIa required neither Zn2+ nor high molecular weight kininogen. CONCLUSIONS: Platelet polyphosphate and Zn2+ can promote subsets of the reactions of the contact pathway, with implications for a variety of disease states.


Asunto(s)
Coagulación Sanguínea , Quininógeno de Alto Peso Molecular/sangre , Polifosfatos/sangre , Zinc/sangre , Activación Enzimática , Factor Xa/metabolismo , Humanos , Cinética , Quininógeno de Alto Peso Molecular/química , Peso Molecular , Polifosfatos/química , Proteolisis
4.
Bioanalysis ; 9(19): 1477-1491, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29056074

RESUMEN

AIM: C1-INH-HAE is caused by activation of plasma kallikrein which subsequently cleaves high-molecular-weight kininogen (HMWK) to generate bradykinin and cHMWK. MATERIALS & METHODS: A novel ion-pair 2D LC-MS/MS assay was developed to measure the 46 kDa cHMWK in plasma as a biomarker for C1-INH-HAE. The sample preparation included sodium dodecyl sulfate denaturation, methanol crash, chymotryptic digestion and peptide enrichment by solid phase extraction. RESULTS: The LLOQ was 200 ng/ml. The overall cHMWK recovery combining crash and digestion was 57.5%. The precision of the method was ≤12.7% and accuracy ≤-13.8%. CONCLUSION: A reagent-free LC-MS assay has been developed for the quantitation of 46 kDa cHMWK, which was shown to be elevated in plasma of C1-INH-HAE patients due to C1-INH deficiency relative to that of healthy subjects.


Asunto(s)
Análisis Químico de la Sangre/métodos , Proteína Inhibidora del Complemento C1/genética , Angioedema Hereditario Tipos I y II/sangre , Angioedema Hereditario Tipos I y II/genética , Quininógeno de Alto Peso Molecular/sangre , Proteolisis , Secuencia de Aminoácidos , Biomarcadores/sangre , Biomarcadores/química , Cromatografía Liquida , Humanos , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/aislamiento & purificación , Quininógeno de Alto Peso Molecular/metabolismo , Extracción en Fase Sólida , Espectrometría de Masas en Tándem
5.
Chem Immunol Allergy ; 100: 205-13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24925400

RESUMEN

The formation of bradykinin in plasma requires interaction of three proteins, namely coagulation factor XII (Hageman factor), prekallikrein and high-molecular-weight kininogen (HK). Prekallikrein and HK circulate as a bimolecular complex. Initiation of the cascade upon binding to negatively charged surfaces (or macromolecules) is dependent on factor XII autoactivation, conversion of prekallikrein to kallikrein, and a feedback activation of factor XII by kallikrein. The latter reaction is extremely rapid relative to factor XII autoactivation. The kallikrein then digests HK to liberate bradykinin. The natural surface appears to be vascular endothelial cells which express binding proteins for factor XII and HK, and activation can proceed along the cell surface. Recent findings demonstrate that prekallikrein has enzymatic activity separate from that of kallikrein such that it can stoichiometrically bind and cleave HK to liberate bradykinin. It is normally prevented from doing so by the plasma C1 inhibitor. Release of heat shock protein 90 (HSP-90) from endothelial cells can convert prekallikrein to kallikrein (stoichiometrically) within the prekallikrein-HK complex, even in the absence of factor XII, and the prekallikrein-HK complex can autoactivate to generate kallikrein if phosphate is the buffering ion. The effects of phosphate ion and HSP-90 are additive. Thus, an active site appears to be induced in prekallikrein by binding to HK and any of the aforementioned reactions can generate kallikrein prior to factor XII activation by autoactivation of the HK-PK complex. This brief review highlights the major discoveries made over the past 50 years which have led to our current concepts regarding the constituents and mechanisms of activation of the plasma bradykinin-forming cascade.


Asunto(s)
Bradiquinina/metabolismo , Bradiquinina/química , Células Endoteliales/metabolismo , Factor XII/química , Factor XII/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Precalicreína/química , Precalicreína/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
6.
Protein Sci ; 23(8): 1013-22, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24810540

RESUMEN

High-molecular-weight kininogen domain 5 (HK5) is an angiogenic modulator that is capable of inhibiting endothelial cell proliferation, migration, adhesion, and tube formation. Ferritin can bind to a histidine-glycine-lysine-rich region within HK5 and block its antiangiogenic effects. However, the molecular intricacies of this interaction are not well understood. Analysis of the structure of HK5 using circular dichroism and nuclear magnetic resonance [(1) H, (15) N]-heteronuclear single quantum coherence determined that HK5 is an intrinsically unstructured protein, consistent with secondary structure predictions. Equilibrium binding studies using fluorescence anisotropy were used to study the interaction between ferritin and HK5. The interaction between the two proteins is mediated by metal ions such as Co(2+) , Cd(2+) , and Fe(2+) . This metal-mediated interaction works independently of the loaded ferrihydrite core of ferritin and is demonstrated to be a surface interaction. Ferritin H and L bind to HK5 with similar affinity in the presence of metals. The ferritin interaction with HK5 is the first biological function shown to occur on the surface of ferritin using its surface-bound metals.


Asunto(s)
Ferritinas/química , Proteínas Intrínsecamente Desordenadas/química , Quininógeno de Alto Peso Molecular/química , Metales Pesados/química , Ferritinas/metabolismo , Humanos , Proteínas Intrínsecamente Desordenadas/aislamiento & purificación , Proteínas Intrínsecamente Desordenadas/metabolismo , Quininógeno de Alto Peso Molecular/aislamiento & purificación , Quininógeno de Alto Peso Molecular/metabolismo , Metales Pesados/metabolismo , Modelos Moleculares
7.
PLoS One ; 7(11): e50399, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226277

RESUMEN

Nucleic acids, especially extracellular RNA, are exposed following tissue- or vessel damage and have previously been shown to activate the intrinsic blood coagulation pathway in vitro and in vivo. Yet, no information on structural requirements for the procoagulant activity of nucleic acids is available. A comparison of linear and hairpin-forming RNA- and DNA-oligomers revealed that all tested oligomers forming a stable hairpin structure were protected from degradation in human plasma. In contrast to linear nucleic acids, hairpin forming compounds demonstrated highest procoagulant activities based on the analysis of clotting time in human plasma and in a prekallikrein activation assay. Moreover, the procoagulant activities of the DNA-oligomers correlated well with their binding affinity to high molecular weight kininogen, whereas the binding affinity of all tested oligomers to prekallikrein was low. Furthermore, four DNA-aptamers directed against thrombin, activated protein C, vascular endothelial growth factor and nucleolin as well as the naturally occurring small nucleolar RNA U6snRNA were identified as effective cofactors for prekallikrein auto-activation. Together, we conclude that hairpin-forming nucleic acids are most effective in promoting procoagulant activities, largely mediated by their specific binding to kininogen. Thus, in vivo application of therapeutic nucleic acids like aptamers might have undesired prothrombotic or proinflammatory side effects.


Asunto(s)
Aptámeros de Nucleótidos/química , Coagulación Sanguínea , Coagulantes/química , Secuencias Invertidas Repetidas , Quininógeno de Alto Peso Molecular/química , Oligorribonucleótidos/química , Pruebas de Coagulación Sanguínea , Humanos , Conformación de Ácido Nucleico , Fosfoproteínas/química , Precalicreína/química , Unión Proteica , Proteína C/química , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , Proteínas de Unión al ARN/química , Relación Estructura-Actividad , Trombina/química , Factor A de Crecimiento Endotelial Vascular/química , Nucleolina
8.
Thromb Haemost ; 105(6): 1053-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21544310

RESUMEN

The physiologic activation of the plasma kallikrein-kinin system requires the assembly of its constituents on a cell membrane. High- molecular-weight kininogen (HK) and cleaved HK (HKa) both interact with at least three endothelial cell binding proteins: urokinase plasminogen activator receptor (uPAR), globular C1q receptor (gC1qR,) and cytokeratin 1 (CK1). The affinity of HK and HKa for endothelial cells are KD=7-52 nM. The contribution of each protein is unknown. We examined the direct binding of HK and HKa to the soluble extracellular form of uPAR (suPAR), gC1qR and CK1 using surface plasmon resonance. Each binding protein linked to a CM-5 chip and the association, dissociation and KD (equilibrium constant) were measured. The interaction of HK and HKa with each binding protein was zinc-dependent. The affinity for HK and HKa was gC1qR>CK1>suPAR, indicating that gC1qR is dominant for binding. The affinity for HKa compared to HK was the same for gC1qR, 2.6-fold tighter for CK1 but 53-fold tighter for suPAR. Complex between binding proteins was only observed between gC1qR and CK1 indicating that a binary CK1-gC1qR complex can form independently of kininogen. Although suPAR has the weakest affinity of the three binding proteins, it is the only one that distinguished between HK and HKa. This finding indicates that uPAR may be a key membrane binding protein for differential binding and signalling between the cleaved and uncleaved forms of kininogen. The role of CK1 and gC1qR may be to initially bind HK to the membrane surface before productive cleavage to HKa.


Asunto(s)
Queratinas/metabolismo , Quininógeno de Alto Peso Molecular/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Resonancia por Plasmón de Superficie , Coagulación Sanguínea , Endotelio/metabolismo , Humanos , Queratina-1/metabolismo , Queratinas/química , Quininógeno de Alto Peso Molecular/química , Glicoproteínas de Membrana/química , Unión Proteica , Receptores de Complemento/química , Receptores del Activador de Plasminógeno Tipo Uroquinasa/química , Transducción de Señal
9.
Thromb Haemost ; 104(5): 875-85, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20886177

RESUMEN

We have shown that cleaved high-molecular-weight kininogen inhibits endothelial cell tube and vacuole formation in a concentration-dependent manner and this correlates with its recognised anti-angiogenic activity. The antibody against the urokinase plasminogen activator receptor (uPAR) mimicked the inhibitory effect of cleaved kininogen (HKa) on apoptosis (HKa: 30% and uPAR antibody: 26%) and tube formation. In tumour angiogenesis, cancer cells release angiogenic stimulators, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), thus stimulating the transformation of endogenous pro-uPA to uPA. The proteolytic enzyme urokinase plasminogen activator (uPA) then binds to its receptor in a complex with its inhibitor PAI-1, which results in the internalisation of this complex, and activates extracellular signal-regulated kinase (ERK). Recycling of the uPAR regulates the migration of endothelial cells (ECs). ERK activation stimulates migration and proliferation and suppresses apoptosis of ECs. HKa disrupted the uPA-uPAR complex, inhibited ERK activation, and blocked the internalization of uPAR, eventually resulting in cell death and cell motility arrest. Both are critical steps in angiogenesis.


Asunto(s)
Diferenciación Celular , Membrana Celular/metabolismo , Células Endoteliales/metabolismo , Quininógeno de Alto Peso Molecular/sangre , Neovascularización Fisiológica , Transducción de Señal , Animales , Apoptosis , Movimiento Celular , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Quininógeno de Alto Peso Molecular/química , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Conformación Proteica , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Relación Estructura-Actividad , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Familia-src Quinasas/metabolismo
10.
Reprod Sci ; 16(12): 1144-52, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19657137

RESUMEN

To date, there is no reliable test to identify women in early pregnancy at risk of developing preeclampsia. Difference gel electrophoresis (DIGE) identified the plasma proteins vitronectin (VN) and high-molecular-weight kininogen (HK) in association with preeclampsia. In a longitudinal proteomics study, the plasma of preeclamptic patients (n = 6) was compared to healthy control participants (n = 6) before the onset of preeclampsia (week 20) and at the time of presentation with clinical disease (weeks 33-36). The 75-kd single-chain VN molecule increased 1.6- to 1.9-fold in preeclampsia, whereas the 65-kd moiety of the 2-chain VN molecule decreased 1.5- to 1.7-fold compared to healthy controls (P < .05). Immunoblots revealed differences in proteolytic processing of VN and/or HK in women who develop preeclampsia or preeclampsia further complicated by small-for-gestational-age. Vitronectin and HK may prove to be useful as early markers of fibrinolytic activity and neutrophil activation, which are known to be associated with preeclampsia.


Asunto(s)
Quininógeno de Alto Peso Molecular/sangre , Preeclampsia/sangre , Procesamiento Proteico-Postraduccional , Vitronectina/sangre , Adulto , Biomarcadores/sangre , Western Blotting , Estudios de Casos y Controles , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Quininógeno de Alto Peso Molecular/química , Estudios Longitudinales , Peso Molecular , Preeclampsia/inmunología , Embarazo , Resultado del Embarazo , Segundo Trimestre del Embarazo , Tercer Trimestre del Embarazo , Índice de Severidad de la Enfermedad , Espectrometría de Masas en Tándem , Vitronectina/química
11.
Oncogene ; 28(30): 2756-65, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19483730

RESUMEN

Upregulation and activation of epidermal growth factor receptor and/or urokinase-type plasminogen activator receptor in a variety of cancers have been shown to be associated with poor prognosis. High-molecular-weight kininogen can be hydrolysed by plasma kallikrein to bradykinin and cleaved high-molecular-weight kininogen (HKa). HKa and its domain 5 (D5) both have been shown to have potent anti-angiogenic activity. We now show that HKa blocks human prostate cancer cell (DU145) migration by 76.0+/-2.4% at 300 nM and invasion by 78.0+/-12.9% at 11.1 nM. D5 inhibits tumor migration and invasion in a concentration-dependent manner. Stimulation by basic fibroblast growth factor (bFGF) or vascular endothelial growth factor results in clustering of urokinase-type plasminogen activator receptor (uPAR) and epidermal growth factor receptor (EGFR) on the surface of DU145 cells. The co-localization of uPAR and EGFR is prevented by HKa. Immunoprecipitation suggests that uPAR, EGFR and alpha5beta1 integrin formed a ternary complex. Immunoblotting shows that HKa significantly decreases the bFGF-transactivated phosphorylation of EGFR at Tyr 1173 between 30 min and 4 h. The phosphorylation of extracellular signal-regulated kinase (ERK) and AKT, which are downstream effectors of EGFR, is also inhibited by HKa. These novel data indicate that HKa and D5 inhibit migration and invasion of human prostate cancer cells through an EGFR/uPAR pathway, suggesting the therapeutic potential of HKa and D5 to decrease metastasis of human prostate cancer.


Asunto(s)
Receptores ErbB/fisiología , Quininógeno de Alto Peso Molecular/farmacología , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/uso terapéutico , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/tratamiento farmacológico , Estructura Terciaria de Proteína , Quinazolinas , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
12.
J Allergy Clin Immunol ; 124(1): 143-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19342086

RESUMEN

BACKGROUND: Bradykinin formation typically requires interaction of Factor XII, prekallikrein (PK), and high-molecular-weight kininogen (HK) with negatively charged exogenous initiators or cell-surface proteins. Approximately 85% of plasma PK circulates as a complex with HK. Nonenzymatic cell-derived initiators, such as heat shock protein 90, can activate the HK-PK complex to generate kallikrein, bradykinin, and cleaved HK, even in the absence of Factor XII. OBJECTIVE: We sought to determine whether PK, without activation to kallikrein, can digest HK to release bradykinin. METHODS: Kallikrein was measured by using a chromogenic assay, and bradykinin levels were determined by ELISA. Cleavage of PK and HK were assessed by SDS-PAGE and Western blot analysis. RESULTS: Cleavage of HK by PK is demonstrated without any conversion of PK to kallikrein. HK cleavage by PK is distinguished from that of kallikrein by the following: (1) stoichiometric activation of HK by PK with release of bradykinin proportional to the PK input; (2) inhibition of PK cleavage of HK by corn trypsin inhibitor, which has no effect on kallikrein; and (3) inhibition of PK cleavage of HK by a peptide derived from HK, which inhibits binding of PK to HK. The same peptide has no effect on kallikrein activation of HK. C1 inhibitor (C1INH), the major control protein of the plasma bradykinin-forming cascade, inhibits PK cleavage of HK. CONCLUSION: PK is an enzyme that can cleave HK to release bradykinin, and this reaction is inhibited by C1INH. This might account, in part, for circulating bradykinin levels and initiation of kinin formation in C1INH deficiency.


Asunto(s)
Proteína Inhibidora del Complemento C1/metabolismo , Factor XII/química , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Precalicreína/metabolismo , Western Blotting , Bradiquinina/química , Catálisis , Electroforesis en Gel de Poliacrilamida , Humanos , Precalicreína/antagonistas & inhibidores , Precalicreína/química
13.
Biochem Biophys Res Commun ; 366(4): 938-43, 2008 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-18083112

RESUMEN

Plasma kallikrein kinin system (KKS) activation along with its cellular receptors expression are increased after injury and in patients with septic shock, hypotensive bacteremia and rhesus monkey infected with Salmonella typhimurium. KKS signaling cascade is activated by activated factor XII (FXIIa, Hageman factor)- and prolylcarboxypeptidase (PRCP)-dependent pathways on endothelial cells. Among the many entities that comprise the KKS, high molecular weight kininogen (HK), a bradykinin precursor, is critical in the assembly and activation of this system. HK is primarily expressed in the liver and secreted into the bloodstream. The activation of the KKS influences the permeability of the endothelium by liberating bradykinin (BK) from HK. BK is a potent inflammatory peptide which stimulates constitutive bradykinin B2 and inducible B1 receptors to release nitric oxide and prostacyclin. Regardless of the triggers, PK can only be activated on HK bound to the artificial negatively charged or to cell membrane surfaces. Since LPS has a negatively charged moiety and the ability to induce inflammatory responses in human, we determined the interaction between LPS and HK. HKH19 (HK cell binding site) and heparin inhibited LPS binding to HK with IC(50)s of 15nM and 20 microg/ml, respectively. C1-inhibitor and N-acetylglucosamine glycan inhibited LPS binding to HK with IC(50)s of about 10 microg/ml and 10mM, respectively. This novel study underscores the implication of HK in infection. We propose that HKH19, heparin, and C1-inhibitor present therapeutic potential for the treatment of sepsis and hypotensive bacteremia.


Asunto(s)
Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Lipopolisacáridos/metabolismo , Sitios de Unión , Biotina/metabolismo , Biotinilación , Carbohidratos/farmacología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Heparina/farmacología , Humanos , Concentración de Iones de Hidrógeno , Cinética , Concentración Osmolar , Factores de Tiempo , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/metabolismo
14.
Exp Cell Res ; 314(4): 774-88, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18062965

RESUMEN

Cleaved high molecular weight kininogen (HKa), as well as its domain 5 (D5), inhibits migration and proliferation induced by angiogenic factors and induces apoptosis in vitro. To study its effect on tube formation we utilized a collagen-fibrinogen, three-dimensional gel, an in vitro model of angiogenesis. HKa, GST-D5 and D5 had a similar inhibitory effect of tube length by 90+/-4.5%, 86+/-5.5% and 77+/-12.9%, respectively. D5-derived synthetic peptides: G440-H455 H475-H485 and G486-K502 inhibited tube length by 51+/-3.7%, 54+/-3.8% and 77+/-1.7%, respectively. By a comparison of its inhibitory potency and its sequences, a functional sequence of HKa was defined to G486-G496. PP2, a Src family kinase inhibitor, prevented tube formation in a dose-dependent manner (100-400 nM), but PP3 at 5 microM, an inactive analogue of PP2, did not. HKa and D5 inhibited Src 416 phosphorylation by 62+/-12.3% and 83+/-6.1%, respectively. The C-terminal Src kinase (Csk) inhibits Src kinase activity. Using a siRNA to Csk, expression of Csk was down-regulated by 86+/-7.0%, which significantly increased tube length by 27+/-5.8%. The addition of HKa and D5 completely blocked this effect. We further showed that HKa inhibited Src family kinase activity by disrupting the complex of uPAR, alphavbeta3 integrin and Src. Our results indicate that the anti-angiogenic effect of HKa and D5 is mediated at least in part through Src family kinases and identify a potential novel target for therapeutic inhibition of neovascularization in cancer and inflammatory arthritis.


Asunto(s)
Endotelio Vascular/citología , Quininógeno de Alto Peso Molecular/farmacología , Neovascularización Fisiológica , Familia-src Quinasas/metabolismo , Adhesión Celular , Células Cultivadas , Colágeno/química , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Matriz Extracelular/química , Fibrinógeno/química , Geles , Humanos , Quininógeno de Alto Peso Molecular/química , Péptidos/farmacología , Estructura Terciaria de Proteína
15.
J Thromb Haemost ; 5(12): 2461-6, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17922805

RESUMEN

BACKGROUND: Prekallikrein (PK) plays a central role in the contact system that activates blood coagulation and is involved in the regulation of blood pressure. OBJECTIVES: To provide three-dimensional structural data for PK and rationalize the molecular basis of substrate recognition and zymogen activation. PATIENTS/METHODS: The PK homology model was constructed using the coagulation factor (F) XI crystal structure as a template with the program SWISS-MODEL. RESULTS: The domain organization of the PK apple domains and serine protease is conserved compared to FXI. Surface charge calculations on the PK model revealed that ligand binding to high-molecular-weight kininogen (HK) is predicted to have two key determinants: a pocket within the apple 2 domain and a basic channel formed at the interface of apple domains 1 and 4. A hereditary mutation resulting in PK deficiency (Gly104Arg) and the Lys140 alpha-kallikrein cleavage site both disrupt HK binding and are shown to map to opposite sides of the apple 2 domain pocket. The model also describes the differences in the apple 4 domain that prevents dimer formation in PK vs. FXI. A C-terminal extension in the PK serine protease domain is described as a potential substrate for prolylcarboxypeptidase. CONCLUSIONS: The interaction between PK and HK is mediated by two discrete surfaces formed by the PK A1, A2 and A4 domains with charge likely to be a critical component of the binding. A novel mode of PK activation is postulated to involve prolylcarboxypeptidase cleaving at the C-terminus rather than the activation loop.


Asunto(s)
Quininógeno de Alto Peso Molecular/química , Modelos Moleculares , Precalicreína/química , Secuencias de Aminoácidos , Sitios de Unión , Carboxipeptidasas/metabolismo , Cristalografía por Rayos X , Activación Enzimática , Factor XI/química , Humanos , Imagenología Tridimensional , Quininógeno de Alto Peso Molecular/metabolismo , Ligandos , Mutación , Precalicreína/genética , Precalicreína/metabolismo , Prolina/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato , Propiedades de Superficie , Tirosina/química
16.
Insect Biochem Mol Biol ; 37(5): 466-77, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17456441

RESUMEN

A new kallikrein-kinin system inhibitor, designated anophensin, was identified in the salivary glands of the malaria vector mosquito, Anopheles stephensi. In vitro reconstitution experiments showed that anophensin inhibits activation of the kallikrein-kinin system by inhibiting the reciprocal activation of factor XII (FXII) and prekallikrein (PK), and subsequent release of bradykinin. Additionally, anophensin inhibits activation of the kallikrein-kinin system on cultured human umbilical vein endothelial cells (HUVECs). Direct binding assays show that this inhibitory effect is due to Zn(2+)-dependent specific binding of anophensin to both FXII and high molecular weight kininogen (HK). Furthermore, anophensin interacts with both the N-terminus of FXII and domain D5 of HK, which are the binding domains for biological activating surfaces. These results suggest that anophensin inhibits activation of the kallikrein-kinin system by interfering with the association of FXII and HK with biological activating surfaces, resulting in the inhibition of bradykinin release in a host animal during insect blood-feeding.


Asunto(s)
Anopheles/metabolismo , Factor XII/antagonistas & inhibidores , Proteínas de Insectos/farmacología , Insectos Vectores/metabolismo , Sistema Calicreína-Quinina/efectos de los fármacos , Quininógeno de Alto Peso Molecular/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bradiquinina/metabolismo , Células Cultivadas , Clonación Molecular , ADN Complementario/química , Factor XII/química , Factor XII/metabolismo , Humanos , Proteínas de Insectos/química , Proteínas de Insectos/metabolismo , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Glándulas Salivales/metabolismo , Alineación de Secuencia , Zinc/metabolismo
17.
Am J Physiol Heart Circ Physiol ; 292(6): H2959-65, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17293494

RESUMEN

High-molecular-weight kininogen (HK) and its domain 3 (D3) exhibit anticoagulant properties and inhibit platelet activation at low thrombin concentration in vitro. We hypothesized that the rapid occlusive thrombosis in HK-deficient (HKd) rats following endothelial injury of the aorta results from enhanced platelet aggregation by thrombin. The effects of D3 (G235-M357) or D3-derived peptides on thrombosis in vivo were tested. D3 and its exon 7C terminal peptide (E7CP, K270-Q292), expressed as glutathione S-transferase (GST) fusion proteins (GST-D3, GST-E7CP), or GST alone, as well as cleaved HK (HKa) or synthetic peptide E7CP, were infused intravenously 10 min before endothelial injury. Blood flow was reduced down to 10% of baseline flow within 28 +/- 5.2 min by a platelet-fibrin thrombus in GST-treated HKd rats compared with >240 min in GST-treated normal HK rats (wild type). GST-D3, GST-E7CP, HKa, or E7CP infusion prolonged the flow time to 233, >240, 223, and >240 min, respectively, in HKd rats. When GST-E7CP was infused 10 min after the injury, blood flow was maintained for >240 min. Thrombin-antithrombin concentrations were elevated by injury in HKd rats receiving GST from 35 to 55 microg/l and decreased with GST-E7CP, HKa, or E7CP reconstitution to 40, 15, and 9 microg/l, respectively. We conclude that HKd rats are prothrombotic and that HKa, kininogen D3, and its fragment E7CP modulate arterial thrombosis after endothelial injury.


Asunto(s)
Aorta/metabolismo , Endotelio Vascular/metabolismo , Fibrinolíticos/metabolismo , Quininógeno de Alto Peso Molecular/metabolismo , Fragmentos de Péptidos/metabolismo , Trombosis/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Antitrombina III , Aorta/efectos de los fármacos , Aorta/lesiones , Aorta/patología , Aorta/fisiopatología , Velocidad del Flujo Sanguíneo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/lesiones , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Fibrina/metabolismo , Fibrinolíticos/química , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Glutatión Transferasa/genética , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/genética , Quininógeno de Alto Peso Molecular/farmacología , Quininógeno de Alto Peso Molecular/uso terapéutico , Masculino , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Péptido Hidrolasas/sangre , Agregación Plaquetaria , Estructura Terciaria de Proteína , Ratas , Ratas Endogámicas Lew/genética , Proteínas Recombinantes de Fusión/metabolismo , Flujo Sanguíneo Regional , Trombina/metabolismo , Trombosis/patología , Trombosis/fisiopatología , Trombosis/prevención & control
18.
EMBO J ; 25(23): 5569-78, 2006 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-17093496

RESUMEN

Activation of the contact system has two classical consequences: initiation of the intrinsic pathway of coagulation, and cleavage of high molecular weight kininogen (HK) leading to the release of bradykinin, a potent proinflammatory peptide. In human plasma, activation of the contact system at the surface of significant bacterial pathogens was found to result in further HK processing and bacterial killing. A fragment comprising the D3 domain of HK is generated, and within this fragment a sequence of 26 amino acids is mainly responsible for the antibacterial activity. A synthetic peptide covering this sequence kills several bacterial species, also at physiological salt concentration, as effectively as the classical human antibacterial peptide LL-37. Moreover, in an animal model of infection, inhibition of the contact system promotes bacterial dissemination and growth. These data identify a novel and important role for the contact system in the defence against invasive bacterial infection.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Inmunidad Innata , Quininógeno de Alto Peso Molecular/farmacología , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Antibacterianos/química , Humanos , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Ratones , Datos de Secuencia Molecular , Péptidos/química , Estructura Terciaria de Proteína
19.
Rev. invest. clín ; 57(6): 802-813, Nov.-Dec. 2005. ilus
Artículo en Español | LILACS | ID: lil-632401

RESUMEN

The plasma kallikrein-kinin system (KKS) participates in the pathogenesis of inflammatory reactions involved in cellular injury, coagulation, fibrinolysis, kinin formation, complement activation, cytokine secretion and release of proteases. It has been shown that KKS activation in the systemic inflammatory response syndrome results in decrease of its component plasma proteins. Similar changes have been documented in diabetes, sepsis, children with vasculitis, allograft rejection, disseminated intravascular coagulation, patients with recurrent pregnancy losses, hereditary angioedema, adult respiratory distress syndrome and coronary artery disease. Direct involvement of the KKS in the pathogenesis of experimental acute arthritis and acute and chronic enterocolitis has been documented by previous studies from our laboratory using experimental animal models. It has been found that in HK deficient Lewis rats, experimental IBD was much less severe. We showed a genetic difference in kininogen structure between resistant Buffalo and susceptible Lewis rats, which results in accelerated cleavage of HK and it is responsible for the susceptibility to the inflammatory process in the Lewis rats. It has been demostrated that therapy with a specific plasma kallikrein inhibitor (P8720) modulated the experimental enterocolitis, arthritis and systemic inflammation. Furthermore, it has been shown that a bradykinin 2 receptor (B2R) antagonist attenuates the inflammatory changes in the same animal model. We have showed that a monoclonal antibody targeting HK decreases angiogénesis and arrests tumor growth in a syngeneic animal model. In summary, these results indicate that the plasma KKS plays a central role in the pathogenesis of chronic intestinal inflammation, arthritis and angiogenesis.


Se ha demostrado la participación del sistema plasmático de kalikreína-kininas (KKS) en el proceso inflamatorio, el cual incluye reacciones de daño celular, coagulación y fibrinólisis, formación de kininas, activación del complemento, secreción de citoquinas y liberación de proteasas. El KKS se encuentra activado en el síndrome de respuesta inflamatoria sistémica con una disminución en la concentración plasmática de las proteínas que lo constituyen. También se ha demostrado una activación similar en la diabetes, choque séptico, vasculitis en infantes, enfermedad injerto-huésped, coagulación intravascular diseminada, pacientes con abortos de repetición, angioedema hereditario, el síndrome de estrés respiratorio del adulto y enfermedad coronaria arterial. Mediante el uso de modelos animales experimentales, nuestro laboratorio ha demostrado una participación directa del KKS en la patogénesis de la artritis experimental aguda y la enterocolitis aguda y crónica. Se ha demostrado que en la rata tipo Lewis, cuando es deficiente de kininógeno de alto peso molecular (HK), la enfermedad inflamatoria intestinal es menos severa comparada con la presentada en ratas con niveles normales de HK como la Buffalo. Nosotros mostramos una diferencia entre el gene que codifica la molécula del kininógeno de la rata tipo Buffalo (resistentes) y Lewis (susceptibles), que resulta en un incremento de la actividad proteolítica de kalikreína sobre su substrato HK, lo cual predispone a las ratas Lewis al desarrollo de la enfermedad inflamatoria crónica. Se ha demostrado una disminución en las manifestaciones inflamatorias sistémicas de la enterocolitis y artritis experimental mediante el uso de un inhibidor específico de la kalikreína (P8720). Además, el antagonista del receptor 2 de la bradikinina (BR2) atenuó los cambios inflamatorios en el mismo modelo animal. Asimismo, se ha demostrado que las ratas Lewis deficientes de kininógeno desarrollaron inflamación intestinal sistémica menos severa. Mediante el uso del anticuerpo monoclonal C11C1 contra HK se logró una disminución de la angiogenesis y, consecuentemente, el crecimiento tumoral. En conclusión, los resultados demuestran que el sistema plasmático de KKS desempeña un papel preponderante en la patogénesis de la artritis reumatoide, la enfermedad intestinal crónica y en el proceso angiogénico.


Asunto(s)
Animales , Ratas , Sistema Calicreína-Quinina/fisiología , Quininógeno de Alto Peso Molecular/fisiología , Neovascularización Fisiológica/fisiología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Artritis Reactiva/fisiopatología , Compuestos de Boro/uso terapéutico , Adhesión Celular/fisiología , Fibrinólisis/fisiología , Predisposición Genética a la Enfermedad , Inflamación/fisiopatología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/fisiopatología , Quininógeno de Alto Peso Molecular/biosíntesis , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/deficiencia , Quininógeno de Alto Peso Molecular/genética , Quininógeno de Alto Peso Molecular/uso terapéutico , Modelos Moleculares , Datos de Secuencia Molecular , Oligopéptidos/uso terapéutico , Peptidoglicano/toxicidad , Polisacáridos Bacterianos/toxicidad , Ratas Endogámicas BUF , Ratas Endogámicas Lew , Relación Estructura-Actividad
20.
Thromb Haemost ; 94(3): 606-14, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16268479

RESUMEN

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.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Quininógeno de Alto Peso Molecular/farmacología , Fragmentos de Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Androstadienos/farmacología , Animales , Bovinos , Células Cultivadas , Cromonas/farmacología , Células Endoteliales/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Quininógeno de Alto Peso Molecular/química , Lisofosfolípidos/farmacología , Morfolinas/farmacología , Fragmentos de Péptidos/química , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Estructura Terciaria de Proteína , Esfingosina/análogos & derivados , Esfingosina/farmacología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/farmacología , Wortmanina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...