Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Intervalo de año de publicación
1.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34948166

RESUMEN

Kininogens are multidomain glycoproteins found in the blood of most vertebrates. High molecular weight kininogen demonstrate both carrier and co-factor activity as part of the intrinsic pathway of coagulation, leading to thrombin generation. Kininogens are the source of the vasoactive nonapeptide bradykinin. To date, attempts to crystallize kininogen have failed, and very little is known about the shape of kininogen at an atomic level. New advancements in the field of cryo-electron microscopy (cryoEM) have enabled researchers to crack the structure of proteins that has been refractory to traditional crystallography techniques. High molecular weight kininogen is a good candidate for structural investigation by cryoEM. The goal of this review is to summarize the findings of kininogen structural studies.


Asunto(s)
Quininógeno de Alto Peso Molecular/genética , Quininógeno de Alto Peso Molecular/metabolismo , Quininógeno de Alto Peso Molecular/fisiología , Animales , Bradiquinina/metabolismo , Microscopía por Crioelectrón/métodos , Humanos , Calicreínas/sangre , Quininógenos/genética , Quininógenos/metabolismo , Quininógenos/fisiología , Relación Estructura-Actividad
2.
Adv Immunol ; 121: 41-89, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24388213

RESUMEN

Binding of negatively charged macromolecules to factor XII induces a conformational change such that it becomes a substrate for trace amounts of activated factor present in plasma (less than 0.01%). As activated factor XII (factor XIIa or factor XIIf) forms, it converts prekallikrein (PK) to kallikrein and kallikrein cleaves high molecular weight kininogen (HK) to release bradykinin. A far more rapid activation of the remaining unactivated factor XII occurs by enzymatic cleavage by kallikrein (kallikrein-feedback) and sequential cleavage yields two forms of activated factor XII; namely, factor XIIa followed by factor XII fragment (factor XIIf). PK circulates bound to HK and binding induces a conformational change in PK so that it acquires enzymatic activity and can stoichiometrically cleave HK to produce bradykinin. This reaction is prevented from occurring in plasma by the presence of C1 inhibitor (C1 INH). The same active site leads to autoactivation of the PK-HK complex to generate kallikrein if a phosphate containing buffer is used. Theoretically, formation of kallikrein by this factor XII-independent route can activate surface-bound factor XII to generate factor XIIa resulting in a marked increase in the rate of bradykinin formation as stoichiometric reactions are replaced by Michaelis-Menton, enzyme-substrate, kinetics. Zinc-dependent binding of the constituents of the bradykinin-forming cascade to the surface of endothelial cells is mediated by gC1qR and bimolecular complexes of gC1qR-cytokeratin 1 and cytokeratin 1-u-PAR (urokinase plasminogen activator receptor). Factor XII and HK compete for binding to free gC1qR (present in excess) while cytokeratin 1-u-PAR preferentially binds factor XII and gC1qR-cytokeratin 1 preferentially binds HK. Autoactivation of factor XII can be initiated as a result of binding to gC1qR but is prevented by C1 INH. Yet stoichiometric activation of PK-HK to yield kallikrein in the absence of factor XII can be initiated by heat shock protein 90 (HSP-90) which forms a zinc-dependent trimolecular complex by binding to HK. Thus, endothelial cell-dependent activation can be initiated by activation of factor XII or by activation of PK-HK. Hereditary angioedema (HAE), types I and II, are due to autosomal dominant mutations of the C1 INH gene. In type I disease, the level of C1 INH protein and function is proportionately low, while type II disease has a normal protein level but diminished function. There is trans-inhibition of the one normal gene so that functional levels are 30% or less and severe angioedema affecting peripheral structures, the gastrointestinal tract, and the larynx results. Prolonged incubation of plasma of HAE patients (but not normal controls) leads to bradykinin formation and conversion of PK to kallikrein which is reversed by reconstitution with C1 INH. The disorder can be treated by C1 INH replacement, inhibition of plasma kallikrein, or blockade at the bradykinin B-2 receptor. A recently described HAE with normal C1 INH (based on inhibition of activated C1s) presents similarly; the defect is not yet clear, however one-third of patients have a mutant factor XII gene. We have shown that this HAE has a defect in bradykinin overproduction whether the factor XII mutation is present or not, that patients' C1 INH is capable of inhibiting factor XIIa and kallikrein (and not just activated C1) but the functional level is approximately 40-60% of normal, and that α2 macroglobulin protein levels are normal. In vitro abnormalities can be suppressed by raising C1 INH to twice normal levels. Finally, aggregated proteins have been shown to activate the bradykinin-forming pathway by catalyzing factor XII autoactivation. Those include the amyloid ß protein of Alzheimer's disease and cryoglobulins. This may represent a new avenue for kinin-dependent research in human disease. In allergy (anaphylaxis; perhaps other mast cell-dependent reactions), the oversulfated proteoglycan of mast cells, liberated along with histamine, also catalyze factor XII autoactivation.


Asunto(s)
Angioedemas Hereditarios/inmunología , Angioedemas Hereditarios/patología , Bradiquinina/metabolismo , Inmunidad Innata , Mediadores de Inflamación/fisiología , Transducción de Señal/inmunología , Angioedemas Hereditarios/etiología , Animales , Bradiquinina/sangre , Bradiquinina/fisiología , Proteína Inhibidora del Complemento C1/fisiología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Factor XII/metabolismo , Factor XII/fisiología , Humanos , Inflamación/sangre , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/sangre , Quininógeno de Alto Peso Molecular/fisiología , Precalicreína/fisiología
3.
Front Biosci ; 13: 4667-86, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508538

RESUMEN

Inflammation is an adaptive response to damage of vascularized tissues, which develops according to a stereotyped sequence governed by the local production of the so-called "chemical mediators of inflammation". Here we review the evidences indicating a role of the plasminogen activation system in the regulation of all the phases of the inflammation process. Plasminogen activation controls the formation of complement anaphylotoxins (responsible for vasodilatation, increase of venular permeability and leukocyte chemotaxis) and of bradykinin (which accounts for vasodilatation, increase of venular permeability and pain) by regulating the plasma contact system. The urokinase plasminogen activator and its cellular receptor, expressed on the surface of human leukocytes, provide a functional unit that, by regulating interaction of leukocytes with extracellular matrix, as well as its degradation, is critical for the migration of leukocytes and for their movement in the damaged tissues. By preventing excess fibrin accumulation in inflamed tissues, the plasminogen activation system also governs the proper evolution of the inflammatory exudates and prevents the possibility of a shift from acute to chronic inflammation.


Asunto(s)
Inflamación/fisiopatología , Activadores Plasminogénicos/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Movimiento Celular/fisiología , Receptores ErbB/fisiología , Exudados y Transudados/fisiología , Fibrina/fisiología , Humanos , Integrinas/fisiología , Quininógeno de Alto Peso Molecular/fisiología , Receptores de Superficie Celular/fisiología , Receptores Acoplados a Proteínas G/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Activador de Tejido Plasminógeno/fisiología
5.
Arterioscler Thromb Vasc Biol ; 27(9): 1968-75, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17585065

RESUMEN

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.


Asunto(s)
Adhesión Celular/fisiología , Células Endoteliales/fisiología , Integrina alfaVbeta3/metabolismo , Quininógeno de Alto Peso Molecular/fisiología , Inhibidores de la Angiogénesis/fisiología , Membrana Celular/metabolismo , Regulación hacia Abajo , Humanos , Venas Umbilicales/fisiología
6.
Thromb Haemost ; 95(2): 312-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16493494

RESUMEN

Recent studies have shown that peptides identified as surface binding regions of high molecular mass kininogen (HK) and factor XII (FXII) inhibit the Zn(2+)-dependent binding of FXII to confluent layers of human umbilical vein endothelial cells (HUVEC). This indicates that negatively charged FXII binding surfaces, such as sulfatides and dextran sulfate, may interfere with the binding of FXII to confluent layer of HUVEC. Upon investigating this hypothesis it was unexpectedly found that sulfatides enhanced a specific binding of FXII to a matrix protein expressed during growth of the endothelial cells and that this binding was independent of the presence of Zn(2+). The function of sulfatides was partly to minimize nonspecific electrostatic binding and partly to induce and enhance autoactivation of FXII generating alphaFXIIa. Western blot analysis of the extracts of the matrix incubated with FXII and sulfatides showed that the binding was specific for alphaFXIIa. The dissociation constant for binding alphaFXIIa was 12.8 +/- 0.4 nM (n = 4). The binding of alphaFXIIa to ECM was mapped to the heavy chain as no binding was observed of the light chain containing the catalytic domain. HK, which previously has been shown to completely abolish the Zn(2+)-dependent binding of FXII to confluent layers of HUVEC, did not inhibit the binding of alphaFXIIa to the matrix but sulfatides enhanced binding of FXII to ECM. This suggests that HK interferes with the binding of FXII to sulfatides and thereby the autoactivation of FXII. Trypsin treatment of the matrix protein completely abolished the binding, and fibronectin but not laminin was found to be a suitable target. The binding of activated FXII to the ECM suggests that FXIIa may be a modulator of cellular adhesion, migration and vascularization.


Asunto(s)
Endotelio Vascular/citología , Proteínas de la Matriz Extracelular/metabolismo , Células Endoteliales/química , Células Endoteliales/metabolismo , Endotelio Vascular/química , Proteínas de la Matriz Extracelular/genética , Factor XII/metabolismo , Factor XIIa , Fibronectinas , Humanos , Quininógeno de Alto Peso Molecular/fisiología , Unión Proteica , Subunidades de Proteína , Electricidad Estática , Sulfoglicoesfingolípidos/farmacología , Venas Umbilicales/citología , Zinc
7.
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
8.
Curr Drug Targets ; 6(5): 551-7, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16026275

RESUMEN

Factor XII, plasma prekallikrein and high molecular weight kininogen were first identified as coagulation proteins in the intrinsic pathway because patients deficient in these proteins had marked prolongation of in vitro surface-activated coagulation time. However, deficiencies of these proteins are not associated with clinical bleeding. Paradoxically, studies suggest that these proteins have anticoagulant and profibrinolytic activities. In fact, association between deficiencies of these proteins and thrombosis has been reported. Also, deficiencies of these proteins, auto-antibodies to these proteins and anti-phospholipid antibodies are frequent hemostatis-related abnormalities found in unexplained recurrent aborters. Recently, evidence has accumulated for the presence of the kallikrein-kininogen-kinin system in the fetoplacental unit. Since contact proteins or kallikrein-kininogen-kinin system may play an important role in pregnancy especially in the fetoplacental unit, deficiencies of these proteins and/or auto-antibodies to these proteins may be associated with pregnancy losses. These possibilities will be reviewed, the functions of the individual components will be summarized, and their role in blood coagulation and pregnancy discussed.


Asunto(s)
Factor XII/fisiología , Quininógenos/fisiología , Complicaciones del Embarazo/fisiopatología , Precalicreína/fisiología , Animales , Femenino , Humanos , Quininógeno de Alto Peso Molecular/fisiología , Calicreína Plasmática/fisiología , Embarazo , Complicaciones Hematológicas del Embarazo/terapia
9.
J Thromb Haemost ; 3(4): 670-6, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15733059

RESUMEN

High-molecular-weight kininogen (HK) is a plasma protein that possesses multiple physiological functions. Originally identified as a precursor of bradykinin, a bioactive peptide that regulates many cardiovascular processes, it is now recognized that HK plays important roles in fibrinolysis, thrombosis, and inflammation. HK binds to endothelial cells where it can be cleaved by plasma kallikrein to release bradykinin (BK). The remaining portion of the molecule, cleaved HK, is designated cleaved high-molecular-weight kininogen or HKa. While BK has been intensively studied, the physiological implication of the generation of HKa is not clear. Recent studies have revealed that HKa inhibits angiogenesis while BK promotes angiogenesis. These findings represent novel functions of the kallikrein-kinin system that have not yet been fully appreciated. In this review, we will briefly discuss the recent progress in the studies of the molecular mechanisms that mediate the antiangiogenic effect of HKa and the proangiogenic activity of BK.


Asunto(s)
Bradiquinina/fisiología , Sistema Calicreína-Quinina , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/fisiología , Neovascularización Fisiológica , Inhibidores de la Angiogénesis/química , Animales , Adhesión Celular , Ciclo Celular , Membrana Celular/metabolismo , Proliferación Celular , Humanos , Ratones , Modelos Biológicos , Neovascularización Patológica , Péptidos/química , Estructura Terciaria de Proteína
10.
Rev Invest Clin ; 57(6): 802-13, 2005.
Artículo en Español | MEDLINE | ID: mdl-16708906

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 angiogenesis 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.


Asunto(s)
Sistema Calicreína-Quinina/fisiología , Quininógeno de Alto Peso Molecular/fisiología , Neovascularización Fisiológica/fisiología , Secuencia de Aminoácidos , Animales , 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 , Ratas Endogámicas BUF , Ratas Endogámicas Lew , Relación Estructura-Actividad
11.
Circ Res ; 94(9): 1227-34, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-15044324

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Quininógeno de Alto Peso Molecular/fisiología , Receptores de Superficie Celular/fisiología , Familia-src Quinasas , Anticuerpos Monoclonales/farmacología , Apoptosis/fisiología , Caveolina 1 , Caveolinas/fisiología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Células Cultivadas , Células Endoteliales , Endotelio Vascular , Proteínas de la Matriz Extracelular/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibronectinas/farmacología , Humanos , Integrina alfa5beta1/fisiología , Integrina alfaVbeta3/fisiología , Quininógeno de Alto Peso Molecular/farmacología , Sustancias Macromoleculares , Fosfatidilinositol Diacilglicerol-Liasa/farmacología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-yes , Receptores de Superficie Celular/efectos de los fármacos , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Venas Umbilicales , Vitronectina/farmacología
12.
J Biol Chem ; 278(46): 45375-81, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-12952972

RESUMEN

Leukocyte-platelet interaction is important in mediating leukocyte adhesion to a thrombus and leukocyte recruitment to a site of vascular injury. This interaction is mediated at least in part by the beta2-integrin Mac-1 (CD11b/CD18) and its counter-receptor on platelets, glycoprotein Ibalpha (GPIbalpha). High molecular weight kininogen (HK) was previously shown to interact with both GPIbalpha and Mac-1 through its domains 3 and 5, respectively. In this study we investigated the ability of HK to interfere with the leukocyte-platelet interaction. In a purified system, HK binding to GPIbalpha was inhibited by HK domain 3 and the monoclonal antibody (mAb) SZ2, directed against the epitope 269-282 of GPIbalpha, whereas mAb AP1, directed to the region 201-268 of GPIbalpha had no effect. In contrast, mAb AP1 inhibited the Mac-1-GPIbalpha interaction. Binding of GPIbalpha to Mac-1 was enhanced 2-fold by HK. This effect of HK was abrogated in the presence of HK domains 3 or 5 or peptides from the 475-497 region of the carboxyl terminus of domain 5 as well as in the presence of mAb SZ2 but not mAb AP1. Whereas no difference in the affinity of the Mac-1-GPIbalpha interaction was observed in the absence or presence of HK, maximal binding of GPIbalpha to Mac-1 doubled in the presence of HK. Moreover, HK/HKa increased the Mac-1-dependent adhesion of myelomonocytic U937 cells and K562 cells transfected with Mac-1 to immobilized GPIbalpha or to GPIbalpha-transfected Chinese hamster ovary cells. Finally, Mac-1-dependent adhesion of neutrophils to surface-adherent platelets was enhanced by HK. Thus, HK can bridge leukocytes with platelets by interacting via its domain 3 with GPIbalpha and via its domain 5 with Mac-1 thereby augmenting the Mac-1-GPIbalpha interaction. These distinct molecular interactions of HK with leukocytes and platelets contribute to the regulation of the adhesive behavior of vascular cells and provide novel molecular targets for reducing atherothrombotic pathologies.


Asunto(s)
Plaquetas/metabolismo , Quininógeno de Alto Peso Molecular/metabolismo , Quininógeno de Alto Peso Molecular/fisiología , Leucocitos/metabolismo , Antígeno de Macrófago-1/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Sitios de Unión , Células CHO , Adhesión Celular , Comunicación Celular , Cricetinae , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Epítopos , Citometría de Flujo , Humanos , Células K562 , Neutrófilos/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Unión Proteica , Estructura Terciaria de Proteína , Transfección , Células U937
13.
Curr Vasc Pharmacol ; 1(1): 59-64, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15320853

RESUMEN

During the past 20 years, the proteins of the "contact system", namely high molecular weight kininogen (HK), kallikrein and Factor XII have been shown to have very little direct impact on hemostasis despite their initial description as initiators of the "intrinsic system". In fact these proteins have rather anticoagulant and profibrinolytic properties. The focus of this review is to summarize the known antithrombotic properties of HK demonstrating its potential application for the novel therapeutic interventions against thromboembolic complications. In particular, HK can inhibit platelet aggregation, as (i) its domain 5 interferes with ligand binding of alphaIIbbeta3-integrins, (ii) its domain 3 blocks thrombin-dependent platelet aggregation by interfering with thrombin binding to the glycoprotein Ib-IX-V complex on platelets, (iii) bradykinin, which is derived upon cleavage of HK, blocks thrombin-induced platelet aggregation, and (iv) HK domain 2 can inhibit the function of platelet calpain. Moreover, HK may have profibrinolytic actions as it can (i) inhibit plasminogen activator inhibitor-1 function and (ii) potentiate prourokinase activation with subsequent pericellular plasmin formation. Indeed, patients lacking circulating HK are at increased risk for thrombosis, and a prothrombotic phenotype was reported for kininogen-deficient rats. All these observations render kininogen antithrombotic, rather than prothrombotic, and the ongoing research aims to develop novel kininogen-related antithrombotic therapies.


Asunto(s)
Fibrinolíticos/farmacología , Quininógeno de Alto Peso Molecular/fisiología , Animales , Fibrinolíticos/química , Humanos , Quininógeno de Alto Peso Molecular/química , Agregación Plaquetaria/efectos de los fármacos , Relación Estructura-Actividad , Tromboembolia/metabolismo , Tromboembolia/prevención & control
14.
Int Immunopharmacol ; 2(13-14): 1841-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12489798

RESUMEN

Understanding the importance and physiologic activity of the plasma kallikrein/kinin system (KKS) has been thwarted by the absence of an inclusive theory for its assembly and activation. The contact activation hypothesis describes the assembly and activation of this system in test tubes and disease states, but not under physiologic circumstances. Recent investigations have indicated a new cohesive hypothesis for understanding physiologic activation of this system. Prekallikrein (PK) and factor XI (FXI) through high molecular weight kininogen (HK) assemble on a co-localized, multiprotein receptor complex on endothelial cells that consists of at least cytokeratin 1 (CKI), gClqR, and urokinase plasminogen activator receptor (muPAR). When assembled on these proteins, prekallikrein becomes activated to kallikrein by the membrane-expressed enzyme prolylcarboxypeptidase (PRCP). Formed kallikrein then activates factor XII (FXII) for amplification of its activation and single chain urokinase. The plasma kallikrein/kinin system may serve as a physiologic counterbalance to the plasma renin angiotensin system (RAS) by lowering blood pressure and preventing thrombosis. Insights into the integrated role of these two systems may afford the development of novel therapeutic drugs to manage hypertension and thrombosis.


Asunto(s)
Endotelio/fisiología , Sistema Calicreína-Quinina/fisiología , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa , Calicreína Plasmática/metabolismo , Animales , Sitios de Unión , Unión Competitiva , Membrana Celular/metabolismo , Membrana Celular/fisiología , Endotelio/citología , Endotelio/metabolismo , Factor XI/metabolismo , Factor XI/fisiología , Factor XII/metabolismo , Factor XII/fisiología , Humanos , Sistema Calicreína-Quinina/efectos de los fármacos , Quininógeno de Alto Peso Molecular/metabolismo , Quininógeno de Alto Peso Molecular/fisiología , Calicreína Plasmática/fisiología , Precalicreína/metabolismo , Precalicreína/fisiología
15.
Arch Pathol Lab Med ; 126(11): 1382-6, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12421145

RESUMEN

OBJECTIVES: To review the literature for conditions, diseases, and disorders that affect activity of the contact factors, and further to review the literature for evidence that less than normal activity of any of the contact factors may be associated with thrombophilia. DATA SOURCES: MEDLINE search for English-language articles published from 1988 to 2001 and pertinent references contained therein, as well as search of references in recent relevant articles and reviews. STUDY SELECTION: Relevant clinical and laboratory information was extracted from selected articles. Meta-analysis was not feasible because of heterogeneity of reports. DATA EXTRACTION AND SYNTHESIS: Evidence for association of altered levels of the contact factors and thrombophilia was sought. A wide variety of disorders is associated with decreased activity of the contact factors; chief among these disorders are liver disease, hepatic immaturity of newborns, the antiphospholipid syndrome, and, for factor XII, being of Asian descent. These disorders are more common than homozygous deficiency. The few series and case reports of thrombophilic events in patients homozygous for deficiency of contact factors are not persuasive enough to support causality. The apparent association between levels consistent with heterozygosity (40%-60% of normal) of any of the contact factors (but especially factor XII) in persons with antiphospholipid antibodies appears to be due to falsely decreased in vitro activity levels of these factors, which are normal on antigenic testing. The apparent association with thrombosis is better explained by the antiphospholipid syndrome than by the modest reduction of the levels of contact factors. CONCLUSIONS: Presently, it is not recommended to measure activity of contact factors during routine evaluation of patients who have suffered venous or arterial thromboembolism or acute coronary syndromes.


Asunto(s)
Factor XIa/fisiología , Trombofilia/diagnóstico , Coagulación Sanguínea/fisiología , Pruebas de Coagulación Sanguínea , Factor XII/fisiología , Deficiencia del Factor XII/diagnóstico , Deficiencia del Factor XII/etiología , Deficiencia del Factor XII/fisiopatología , Humanos , Quininógeno de Alto Peso Molecular/deficiencia , Quininógeno de Alto Peso Molecular/fisiología , Guías de Práctica Clínica como Asunto , Precalicreína/deficiencia , Precalicreína/fisiología , Trombofilia/complicaciones , Trombofilia/fisiopatología
16.
Sheng Li Ke Xue Jin Zhan ; 31(1): 7-12, 2000 Jan.
Artículo en Chino | MEDLINE | ID: mdl-12532760

RESUMEN

Current concept of blood coagulation is divided into two stages: an "initiation" stage which is handled by tissue factor pathway, and an "augmentation" stage handled by intrinsic pathway beginning in factor XI. Recent studies have demonstrated that the contact system is a modulator for vascular biology with vascular tone regulation, anticoagulant, profibrinolytic, antiadhesive and proinflammatory functions. Changes of contact system are associated with sepsis, thrombosis, etc.


Asunto(s)
Coagulación Sanguínea/fisiología , Quininógeno de Alto Peso Molecular/fisiología , Precalicreína/fisiología , Animales , Factor XII/fisiología , Factor XIIa/fisiología , Humanos , Tromboplastina/fisiología
18.
J Theor Biol ; 191(2): 213-9, 1998 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-9631566

RESUMEN

A mathematical model of contact activation of blood coagulation was developed and analysed. The model variables are concentrations of factor XIIa, kallikrein and activated high-molecular-weight kininogen. Concentrations of active factors were shown to depend on the activating signal value in a hysteretic manner. Within a range of relatively small signals, two (activated and non-activated) stable states coexist (bistability). Signals of the natural environment (surfaces of endothelial and blood cells) seem to be in the range of bistability; therefore, contact activation that persists for a short time can induce a transition of the system to the activated state, and, correspondingly, the formation of a clot. The system cannot return to the initial state, which is characterized by low activation levels, until the activating signals decrease significantly below those present in the circulation.


Asunto(s)
Coagulación Sanguínea , Modelos Biológicos , Factor XIIa/fisiología , Hemorreología , Humanos , Calicreínas/fisiología , Quininógeno de Alto Peso Molecular/fisiología , Matemática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...