Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 8: 14151, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28218242

RESUMEN

Protein disulfide isomerase (PDI), secreted by platelets and endothelial cells on vascular injury, is required for thrombus formation. Using PDI variants that form mixed disulfide complexes with their substrates, we identify by kinetic trapping multiple substrate proteins, including vitronectin. Plasma vitronectin does not bind to αvß3 or αIIbß3 integrins on endothelial cells and platelets. The released PDI reduces disulfide bonds on plasma vitronectin, enabling vitronectin to bind to αVß3 and αIIbß3. In vivo studies of thrombus generation in mice demonstrate that vitronectin rapidly accumulates on the endothelium and the platelet thrombus following injury. This process requires PDI activity and promotes platelet accumulation and fibrin generation. We hypothesize that under physiologic conditions in the absence of secreted PDI, thrombus formation is suppressed and maintains a quiescent, patent vasculature. The release of PDI during vascular injury may serve as a regulatory switch that allows activation of proteins, among them vitronectin, critical for thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Células Endoteliales/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/metabolismo , Lesiones del Sistema Vascular/metabolismo , Animales , Células Cultivadas , Endotelio/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Ratones Noqueados , Mutación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Proteína Disulfuro Isomerasas/genética , Trombosis/genética , Lesiones del Sistema Vascular/genética , Vitronectina/genética , Vitronectina/metabolismo
2.
Antioxid Redox Signal ; 24(1): 1-15, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26467859

RESUMEN

SIGNIFICANCE: The mammalian endoplasmic reticulum (ER) houses a large family of twenty thioredoxin-like proteins of which protein disulfide isomerase (PDI) is the archetypal member. Although the PDI family is best known for its role in oxidative protein folding of secretory proteins in the ER, these thioredoxin-like proteins fulfill ever-expanding roles, both within the secretory pathway and beyond. RECENT ADVANCES: Secreted PDI family proteins have now been shown to serve a critical role in platelet thrombus formation and fibrin generation. Utilizing intravital microscopy to visualize thrombus formation in mice, we have demonstrated the presence of extracellular PDI antigen during thrombus formation following injury of the vascular wall. Inhibition of PDI abrogates thrombus formation in vivo (16, 26, 46, 55). These observations have been extended to other PDI family members, including ERp57 (39, 116, 118, 123) and ERp5 (77). The vascular thiol isomerases are those PDI family members secreted from platelets and/or endothelium (40): PDI, ERp57, ERp5, ERp72, ERp44, ERp29, and TMX3. We focus here on PDI (16, 46, 55), ERp57 (39, 116, 118, 123), and ERp5 (77), which have been implicated in thrombus formation in vivo. CRITICAL ISSUES: It would appear that a system of thiol isomerase redox catalysts has been hijacked from the ER to regulate thrombus formation in the vasculature. FUTURE DIRECTIONS: How this redox system is trafficked to and regulated at the cell surface, the identity of extracellular substrates, why so many thiol isomerases are required, and which thiol isomerase functions are necessary are critical unanswered questions in understanding the role of thiol isomerases in thrombus formation.


Asunto(s)
Proteína Disulfuro Isomerasas/metabolismo , Trombosis/metabolismo , Animales , Humanos , Integrina beta3/metabolismo , Oxidación-Reducción , Unión Proteica
3.
J Biol Chem ; 290(39): 23543-52, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26240139

RESUMEN

Quercetin-3-rutinoside inhibits thrombus formation in a mouse model by inhibiting extracellular protein disulfide isomerase (PDI), an enzyme required for platelet thrombus formation and fibrin generation. Prior studies have identified PDI as a potential target for novel antithrombotic agents. Using a fluorescence enhancement-based assay and isothermal calorimetry, we show that quercetin-3-rutinoside directly binds to the b' domain of PDI with a 1:1 stoichiometry. The binding of quercetin-3-rutinoside to PDI induces a more compact conformation and restricts the conformational flexibility of PDI, as revealed by small angle x-ray scattering. The binding sites of quercetin-3-rutinoside to PDI were determined by studying its interaction with isolated fragments of PDI. Quercetin-3-rutinoside binds to the b'x domain of PDI. The infusion of the b'x fragment of PDI rescued thrombus formation that was inhibited by quercetin-3-rutinoside in a mouse thrombosis model. This b'x fragment does not possess reductase activity and, in the absence of quercetin-3-rutinoside, does not affect thrombus formation in vivo. The isolated b' domain of PDI has potential as an antidote to reverse the antithrombotic effect of quercetin-3-rutinoside by binding and neutralizing quercetin-3-rutinoside.


Asunto(s)
Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Rutina/farmacología , Animales , Sitios de Unión , Calorimetría , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Proteína Disulfuro Isomerasas/metabolismo , Rutina/metabolismo , Dispersión del Ángulo Pequeño , Trombosis/prevención & control , Difracción de Rayos X
4.
Nat Chem Biol ; 11(4): 292-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25686372

RESUMEN

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond. These proteins include numerous bacterial virulence factors, and thus bacterial enzymes that promote disulfide bond formation represent targets for compounds inhibiting bacterial virulence. Here, we describe a new target- and cell-based screening methodology for identifying compounds that inhibit the disulfide bond-forming enzymes Escherichia coli DsbB (EcDsbB) or Mycobacterium tuberculosis VKOR (MtbVKOR), which can replace EcDsbB, although the two are not homologs. Initial screening of 51,487 compounds yielded six specifically inhibiting EcDsbB. These compounds share a structural motif and do not inhibit MtbVKOR. A medicinal chemistry approach led us to select related compounds, some of which are much more effective DsbB inhibitors than those found in the screen. These compounds inhibit purified DsbB and prevent anaerobic growth of E. coli. Furthermore, these compounds inhibit all but one of the DsbBs of nine other Gram-negative pathogenic bacteria tested.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Escherichia coli/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Mycobacterium tuberculosis/metabolismo , Agar/química , Antibacterianos/química , Dominio Catalítico , Química Farmacéutica/métodos , Técnicas Químicas Combinatorias , Disulfuros , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Transporte de Electrón , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/química , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Mycobacterium smegmatis/metabolismo , Conformación Proteica , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/química , Pseudomonas aeruginosa/metabolismo
5.
Blood ; 125(10): 1633-42, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25593336

RESUMEN

Protein disulfide isomerase (PDI), secreted from platelets and endothelial cells after injury, is required for thrombus formation. The effect of platelet and endothelial cell granule contents on PDI-mediated thrombus formation was studied by intravital microscopy using a mouse model of Hermansky-Pudlak syndrome in which platelet dense granules are absent. Platelet deposition and fibrin generation were nearly absent, and extracellular PDI was significantly reduced in HPS6(-/-) mice after vascular injury. HPS6(-/-) platelets displayed impaired PDI secretion and impaired exocytosis of α granules, lysosomes, and T granules due to decreased sensitivity to thrombin, but these defects could be corrected by addition of subthreshold amounts of adenosine 5'-diphosphate (ADP). Human Hermansky-Pudlak syndrome platelets demonstrated similar characteristics. Infusion of wild-type platelets rescued thrombus formation in HPS6(-/-) mice. Human umbilical vein endothelial cells in which the HPS6 gene was silenced displayed impaired PDI secretion and exocytosis of Weibel-Palade bodies. Defective thrombus formation in Hermansky-Pudlak syndrome, associated with impaired exocytosis of residual granules in endothelial cells and platelets, the latter due to deficiency of ADP, is characterized by a defect in T granule secretion, a deficiency in extracellular PDI secretion, and impaired fibrin generation and platelet aggregation. Hermansky-Pudlak syndrome is an example of a hereditary disease whereby impaired PDI secretion contributes to a bleeding phenotype.


Asunto(s)
Plaquetas/enzimología , Células Endoteliales/enzimología , Síndrome de Hermanski-Pudlak/sangre , Síndrome de Hermanski-Pudlak/enzimología , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/sangre , Trombosis/enzimología , Adenosina Difosfato/deficiencia , Adenosina Difosfato/metabolismo , Adenosina Difosfato/farmacología , Animales , Apirasa/metabolismo , Apirasa/farmacología , Plaquetas/efectos de los fármacos , Degranulación de la Célula , Modelos Animales de Enfermedad , Células Endoteliales/patología , Exocitosis/efectos de los fármacos , Femenino , Fibrina/biosíntesis , Síndrome de Hermanski-Pudlak/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Agregación Plaquetaria , Proteína Disulfuro Isomerasas/sangre , ARN Interferente Pequeño/genética , Trombina/metabolismo , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética
6.
Blood ; 125(14): 2276-85, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25624318

RESUMEN

Protein disulfide isomerase (PDI) and endoplasmic reticulum protein 57 (ERp57) are emerging as important regulators of thrombus formation. Another thiol isomerase, endoplasmic reticulum protein 5 (ERp5), is involved in platelet activation. We show here the involvement of ERp5 in thrombus formation using the mouse laser-injury model of thrombosis and a specific antibody raised against recombinant ERp5. Anti-ERp5 antibody inhibited ERp5-dependent platelet and endothelial cell disulfide reductase activity in vitro. ERp5 release at the thrombus site was detected after infusion of Alexa Fluor 488-labeled anti-ERp5 antibody at 0.05 µg/g body weight, a dose that does not inhibit thrombus formation. Anti-ERp5 at 3 µg/g body weight inhibited laser-induced thrombus formation in vivo by causing a 70% decrease in the deposition of platelets and a 62% decrease in fibrin accumulation compared to infusion of control antibody (P < .01). ERp5 binds to ß3 integrin with an equilibrium dissociation constant (KD) of 21 µM, measured by surface plasmon resonance. The cysteine residues in the ERp5 active sites are not required for binding to ß3 integrin. These results provide evidence for a novel role of ERp5 in thrombus formation, a function that may be mediated through its association with αIIbß3.


Asunto(s)
Modelos Animales de Enfermedad , Integrina beta3/metabolismo , Rayos Láser/efectos adversos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/patología , Animales , Plaquetas/metabolismo , Plaquetas/patología , Western Blotting , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Ensayo de Inmunoadsorción Enzimática , Fibrina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Activación Plaquetaria , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Trombosis/enzimología , Trombosis/etiología
7.
Blood ; 124(4): 611-22, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24825863

RESUMEN

Antiphospholipid syndrome (APS) is defined by thrombosis, fetal loss, and the presence of antiphospholipid antibodies, including anti-ß2-glycoprotein-1 autoantibodies (anti-ß2GP1) that have a direct role in the pathogenesis of thrombosis in vivo. The cellular targets of the anti-ß2GP1 autoantibody/ß2GP1 complex in vivo were studied using a laser-induced thrombosis model of APS in a live mouse and human anti-ß2GP1 autoantibodies affinity-purified from APS patients. Cell binding of fluorescently labeled ß2GP1 and anti-ß2GP1 autoantibodies revealed their colocalization on the platelet thrombus but not the endothelium. Anti-ß2GP1 autoantibodies enhanced platelet activation, monitored by calcium mobilization, and endothelial activation, monitored by intercellular adhesion molecule-1 expression. When eptifibatide was infused to block platelet thrombus formation, enhanced fibrin generation and endothelial cell activation were eliminated. Thus, the anti-ß2GP1 autoantibody/ß2GP1 complex binds to the thrombus, enhancing platelet activation, and platelet secretion leads to enhanced endothelium activation and fibrin generation. These results lead to a paradigm shift away from the concept that binding of the anti-ß2GP1 autoantibody/ß2GP1 complex activates both endothelial cells and platelets toward one in which activation of platelets in response to anti-ß2GP1 autoantibody/ß2GP1 complex binding leads to subsequent enhanced endothelium activation and fibrin generation.


Asunto(s)
Anticuerpos Antifosfolípidos/sangre , Síndrome Antifosfolípido/inmunología , Plaquetas/inmunología , Modelos Animales de Enfermedad , Endotelio/inmunología , Trombosis/inmunología , beta 2 Glicoproteína I/metabolismo , Animales , Síndrome Antifosfolípido/metabolismo , Síndrome Antifosfolípido/patología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Plaquetas/citología , Plaquetas/metabolismo , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Endotelio/citología , Endotelio/metabolismo , Fibrina/metabolismo , Fibrinógeno/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Activación Plaquetaria , Trombosis/metabolismo , Trombosis/patología , beta 2 Glicoproteína I/inmunología
8.
Thromb Res ; 130 Suppl 1: S44-6, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23026660

RESUMEN

An electron transport system regulates the initiation of thrombus formation through the activation of critical receptors involved in hemostasis and thrombosis. Protein disulfide isomerase along with other thiol isomerases, important for intracellular protein synthesis, are responsible for this extracellular activity during thrombus formation. Inhibition of these thiol isomerases blocks platelet aggregation and fibrin generation. Pharmaceuticals directed against these thiol isomerases offers a novel approach to antithrombotic therapy.


Asunto(s)
Plaquetas/enzimología , Fibrina/metabolismo , Proteína Disulfuro Isomerasas/sangre , Trombina/metabolismo , Trombosis/sangre , Animales , Plaquetas/efectos de los fármacos , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Fibrinolíticos/farmacología , Humanos , Cinética , Microscopía Confocal , Microscopía por Video , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Trombosis/tratamiento farmacológico , Trombosis/enzimología
9.
Blood ; 120(3): 647-55, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22653978

RESUMEN

Extracellular protein disulfide isomerase (PDI) is required for platelet thrombus formation and fibrin generation after arteriolar wall injury in live mice. PDI is secreted from platelets and endothelial cells on cellular activation, but the mechanism of capture of secreted PDI within the injured vasculature is unknown. We establish that, like the endothelial ß3 integrin α(V)ß(3), the platelet integrin α(IIb)ß(3) binds PDI. PDI also binds to recombinant ß3. Using intravital microscopy, we demonstrate that PDI accumulation at the site of laser-induced arteriolar wall injury is markedly reduced in ß3-null (ß3(-/-)) mice, and neither a platelet thrombus nor fibrin is generated at the vessel injury site. The absence of fibrin after vascular injury in ß3(-/-) mice is because of the absence of extracellular PDI. To evaluate the relative importance of endothelial α(V)ß(3) versus platelet α(IIb)ß(3) or α(V)ß(3), we performed reciprocal bone marrow transplants on wild-type and ß3(-/-) mice. PDI accumulation and platelet thrombus formation were markedly decreased after vessel injury in wild-type mice transplanted with ß3(-/-) bone marrow or in ß3(-/-) mice transplanted with wild-type bone marrow. These results indicate that both endothelial and platelet ß3 integrins contribute to extracellular PDI binding at the vascular injury site.


Asunto(s)
Plaquetas/enzimología , Células Endoteliales/enzimología , Integrina beta3/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/metabolismo , Animales , Arteriolas/enzimología , Arteriolas/lesiones , Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Trasplante de Médula Ósea , Células CHO , Cricetinae , Espacio Extracelular/enzimología , Integrina alfaVbeta3/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía por Video , Músculo Esquelético/irrigación sanguínea , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Quimera por Trasplante
10.
J Clin Invest ; 122(6): 2104-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22565308

RESUMEN

Thrombosis, or blood clot formation, and its sequelae remain a leading cause of morbidity and mortality, and recurrent thrombosis is common despite current optimal therapy. Protein disulfide isomerase (PDI) is an oxidoreductase that has recently been shown to participate in thrombus formation. While currently available antithrombotic agents inhibit either platelet aggregation or fibrin generation, inhibition of secreted PDI blocks the earliest stages of thrombus formation, suppressing both pathways. Here, we explored extracellular PDI as an alternative target of antithrombotic therapy. A high-throughput screen identified quercetin-3-rutinoside as an inhibitor of PDI reductase activity in vitro. Inhibition of PDI was selective, as quercetin-3-rutinoside failed to inhibit the reductase activity of several other thiol isomerases found in the vasculature. Cellular assays showed that quercetin-3-rutinoside inhibited aggregation of human and mouse platelets and endothelial cell-mediated fibrin generation in human endothelial cells. Using intravital microscopy in mice, we demonstrated that quercetin-3-rutinoside blocks thrombus formation in vivo by inhibiting PDI. Infusion of recombinant PDI reversed the antithrombotic effect of quercetin-3-rutinoside. Thus, PDI is a viable target for small molecule inhibition of thrombus formation, and its inhibition may prove to be a useful adjunct in refractory thrombotic diseases that are not controlled with conventional antithrombotic agents.


Asunto(s)
Plaquetas/metabolismo , Fibrinolíticos/farmacología , Agregación Plaquetaria/efectos de los fármacos , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Rutina/farmacología , Trombosis/tratamiento farmacológico , Animales , Inhibidores Enzimáticos/farmacología , Fibrina/genética , Fibrina/metabolismo , Humanos , Ratones , Proteína Disulfuro Isomerasas/efectos adversos , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/farmacología , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Trombosis/inducido químicamente , Trombosis/enzimología
11.
J Clin Invest ; 122(2): 558-68, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22214850

RESUMEN

Hypercholesterolemia is a major risk factor for atherosclerosis. It also is associated with platelet hyperactivity, which increases morbidity and mortality from cardiovascular disease. However, the mechanisms by which hypercholesterolemia produces a procoagulant state remain undefined. Atherosclerosis is associated with accumulation of oxidized lipoproteins within atherosclerotic lesions. Small quantities of oxidized lipoproteins are also present in the circulation of patients with coronary artery disease. We therefore hypothesized that hypercholesterolemia leads to elevated levels of oxidized LDL (oxLDL) in plasma and that this induces expression of the procoagulant protein tissue factor (TF) in monocytes. In support of this hypothesis, we report here that oxLDL induced TF expression in human monocytic cells and monocytes. In addition, patients with familial hypercholesterolemia had elevated levels of plasma microparticle (MP) TF activity. Furthermore, a high-fat diet induced a time-dependent increase in plasma MP TF activity and activation of coagulation in both LDL receptor-deficient mice and African green monkeys. Genetic deficiency of TF in bone marrow cells reduced coagulation in hypercholesterolemic mice, consistent with a major role for monocyte-derived TF in the activation of coagulation. Similarly, a deficiency of either TLR4 or TLR6 reduced levels of MP TF activity. Simvastatin treatment of hypercholesterolemic mice and monkeys reduced oxLDL, monocyte TF expression, MP TF activity, activation of coagulation, and inflammation, without affecting total cholesterol levels. Our results suggest that the prothrombotic state associated with hypercholesterolemia is caused by oxLDL-mediated induction of TF expression in monocytes via engagement of a TLR4/TLR6 complex.


Asunto(s)
Anticolesterolemiantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Hipercolesterolemia/sangre , Monocitos/metabolismo , Simvastatina/farmacología , Tromboplastina/metabolismo , Animales , Coagulación Sanguínea/fisiología , Células Cultivadas , Chlorocebus aethiops , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Receptores de LDL/genética , Receptores de LDL/metabolismo , Trombosis , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 6/genética , Receptor Toll-Like 6/metabolismo
12.
Methods Mol Biol ; 788: 127-39, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22130705

RESUMEN

Platelet microparticles are submicron vesicles that can support thrombin generation on externalized negatively charged phospholipids. Increased numbers of circulating platelet microparticles have been investigated as the basis of hypercoagulability in a variety of prothrombotic conditions. Measurement of platelet microparticles is not standardized and a number of preanalytic considerations can influence accurate analysis. We describe methodology for light scatter-based flow cytometry as well as impedance-based flow cytometry for the enumeration and characterization of platelet microparticles.


Asunto(s)
Plaquetas/citología , Micropartículas Derivadas de Células , Citometría de Flujo/métodos , Humanos
13.
J Biol Chem ; 286(26): 23345-56, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531712

RESUMEN

Mouse and human prothrombin (ProT) active site specifically labeled with D-Phe-Pro-Arg-CH(2)Cl (FPR-ProT) inhibited tissue factor-initiated thrombin generation in platelet-rich and platelet-poor mouse and human plasmas. FPR-prethrombin 1 (Pre 1), fragment 1 (F1), fragment 1.2 (F1.2), and FPR-thrombin produced no significant inhibition, demonstrating the requirement for all three ProT domains. Kinetics of inhibition of ProT activation by the inactive ProT(S195A) mutant were compatible with competitive inhibition as an alternate nonproductive substrate, although FPR-ProT deviated from this mechanism, implicating a more complex process. FPR-ProT exhibited ∼10-fold more potent anticoagulant activity compared with ProT(S195A) as a result of conformational changes in the ProT catalytic domain that induce a more proteinase-like conformation upon FPR labeling. Unlike ProT and ProT(S195A), the pathway of FPR-ProT cleavage by prothrombinase was redirected from meizothrombin toward formation of the FPR-prethrombin 2 (Pre 2)·F1.2 inhibitory intermediate. Localization of ProT labeled with Alexa Fluor® 660 tethered through FPR-CH(2)Cl ([AF660]FPR-ProT) during laser-induced thrombus formation in vivo in murine arterioles was examined in real time wide-field and confocal fluorescence microscopy. [AF660]FPR-ProT bound rapidly to the vessel wall at the site of injury, preceding platelet accumulation, and subsequently to the thrombus proximal, but not distal, to the vessel wall. [AF660]FPR-ProT inhibited thrombus growth, whereas [AF660]FPR-Pre 1, lacking the F1 membrane-binding domain did not bind or inhibit. Labeled F1.2 localized similarly to [AF660]FPR-ProT, indicating binding to phosphatidylserine-rich membranes, but did not inhibit thrombosis. The studies provide new insight into the mechanism of ProT activation in vivo and in vitro, and the properties of a unique exosite-directed prothrombinase inhibitor.


Asunto(s)
Dominio Catalítico , Protrombina/metabolismo , Tromboplastina/metabolismo , Trombosis/enzimología , Sustitución de Aminoácidos , Animales , Coagulación Sanguínea , Activación Enzimática/genética , Humanos , Cinética , Ratones , Mutación Missense , Estructura Terciaria de Proteína , Protrombina/química , Protrombina/genética , Tromboplastina/química , Tromboplastina/genética , Trombosis/genética
14.
Arterioscler Thromb Vasc Biol ; 31(4): 728-33, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21252066

RESUMEN

Blood microparticles are vesicular structures with a diameter of 100 to 1000 nm that are present in the blood of normal subjects and in patients with various diseases. These microparticles are derived from cells that circulate in the blood and cells associated with the blood vessel wall. Microparticle membranes retain the protein receptors of their parent cells and may retain RNAs and other cytosolic content. On the basis of surface protein expression, microparticles are known to be derived from platelets, granulocytes, monocytes, endothelial cells, smooth muscle cells, and tumor cells. Only a subpopulation of these microparticles expresses tissue factor.


Asunto(s)
Coagulación Sanguínea , Micropartículas Derivadas de Células/metabolismo , Tromboplastina/metabolismo , Trombosis/sangre , Animales , Humanos , Tamaño de la Partícula
15.
Blood ; 117(12): 3453-9, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21245481

RESUMEN

Antiphospholipid syndrome is characterized by thrombosis, recurrent fetal loss, and the presence of the lupus anticoagulant, anticardiolipin antibodies, or anti-ß(2)-glycoprotein-1 (anti-ß(2)-GP1) antibodies. Although anti-ß(2)-GP1 antibodies have been documented as a biomarker for diagnosis of antiphospholipid syndrome, their direct role in the pathogenesis of thrombosis is unknown. We have demonstrated using intravital microscopy that anti-ß(2)-GP1 autoantibodies purified from the sera of patients with antiphospholipid syndrome complicated by thrombosis greatly amplify thrombus size after laser-induced vessel wall injury in live mice. Anti-ß(2)-GP1 autoantibodies from 3 patients with antiphospholipid syndrome were affinity-purified using human ß(2)-GP1 bound to agarose. The effects of purified anti-ß(2)-GP1 IgG autoantibodies, of anti-ß(2)-GP1-depleted IgG, and of IgG from normal human sera on thrombus formation were measured in mice after arterial injury in the cremaster muscle. Before injury, purified anti-ß(2)-GP1 IgG autoantibodies, anti-ß(2)-GP1 antibody-depleted IgG, or IgG from normal human sera were infused. Increasing amounts of purified anti-ß(2)-GP1 autoantibodies increased thrombus size in a dose-dependent manner, whereas neither anti-ß(2)-GP1 antibody-depleted IgG nor IgG from normal serum affected thrombus size. These results indicate that anti-ß(2)-GP1 IgG autoantibodies in antiphospholipid syndrome patient sera are not only a marker of antiphospholipid syndrome but are directly involved in the pathogenesis of thrombosis.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Autoanticuerpos/efectos adversos , Modelos Animales de Enfermedad , Trombosis/etiología , beta 2 Glicoproteína I/inmunología , Adulto , Animales , Síndrome Antifosfolípido/sangre , Arterias/patología , Autoanticuerpos/aislamiento & purificación , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Trombosis/metabolismo , Trombosis/patología , beta 2 Glicoproteína I/metabolismo
16.
J Biol Chem ; 286(9): 7027-32, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21199867

RESUMEN

Plasminogen activator inhibitor-1 (PAI-1), together with its physiological target urokinase-type plasminogen activator (uPA), plays a pivotal role in fibrinolysis, cell migration, and tissue remodeling and is currently recognized as being among the most extensively validated biological prognostic factors in several cancer types. PAI-1 specifically and rapidly inhibits uPA and tissue-type PA (tPA). Despite extensive structural/functional studies on these two reactions, the underlying structural mechanism has remained unknown due to the technical difficulties of obtaining the relevant structures. Here, we report a strategy to generate a PAI-1·uPA(S195A) Michaelis complex and present its crystal structure at 2.3-Å resolution. In this structure, the PAI-1 reactive center loop serves as a bait to attract uPA onto the top of the PAI-1 molecule. The P4-P3' residues of the reactive center loop interact extensively with the uPA catalytic site, accounting for about two-thirds of the total contact area. Besides the active site, almost all uPA exosite loops, including the 37-, 60-, 97-, 147-, and 217-loops, are involved in the interaction with PAI-1. The uPA 37-loop makes an extensive interaction with PAI-1 ß-sheet B, and the 147-loop directly contacts PAI-1 ß-sheet C. Both loops are important for initial Michaelis complex formation. This study lays down a foundation for understanding the specificity of PAI-1 for uPA and tPA and provides a structural basis for further functional studies.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/química , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Dominio Catalítico , Cristalografía , Activación Enzimática/fisiología , Humanos , Mutación , Pichia/genética , Inhibidor 1 de Activador Plasminogénico/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Activador de Plasminógeno de Tipo Uroquinasa/genética
17.
Blood ; 116(22): 4675-83, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20675401

RESUMEN

Laser-induced vessel wall injury leads to rapid thrombus formation in an animal thrombosis model. The target of laser injury is the endothelium. We monitored calcium mobilization to assess activation of the laser-targeted cells. Infusion of Fluo-4 AM, a calcium-sensitive fluorochrome, into the mouse circulation resulted in dye uptake in the endothelium and circulating hematopoietic cells. Laser injury in mice treated with eptifibatide to inhibit platelet accumulation resulted in rapid calcium mobilization within the endothelium. Calcium mobilization correlated with the secretion of lysosomal-associated membrane protein 1, a marker of endothelium activation. In the absence of eptifibatide, endothelium activation preceded platelet accumulation. Laser activation of human umbilical vein endothelial cells loaded with Fluo-4 resulted in a rapid increase in calcium mobilization associated cell fluorescence similar to that induced by adenosine diphosphate (10 µM) or thrombin (1 U/mL). Laser activation of human umbilical vein endothelial cells in the presence of corn trypsin inhibitor treated human plasma devoid of platelets and cell microparticles led to fibrin formation that was inhibited by an inhibitory monoclonal anti-tissue factor antibody. Thus laser injury leads to rapid endothelial cell activation. The laser activated endothelial cells can support formation of tenase and prothrombinase and may be a source of activated tissue factor as well.


Asunto(s)
Calcio/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/lesiones , Fibrina/metabolismo , Trombosis , Animales , Plaquetas/metabolismo , Línea Celular , Células Endoteliales/efectos de la radiación , Endotelio Vascular/metabolismo , Endotelio Vascular/efectos de la radiación , Humanos , Rayos Láser , Ratones , Ratones Endogámicos C57BL
18.
Blood ; 116(22): 4665-74, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20668226

RESUMEN

Protein disulfide isomerase (PDI) catalyzes the oxidation reduction and isomerization of disulfide bonds. We have previously identified an important role for extracellular PDI during thrombus formation in vivo. Here, we show that endothelial cells are a critical cellular source of secreted PDI, important for fibrin generation and platelet accumulation in vivo. Functional PDI is rapidly secreted from human umbilical vein endothelial cells in culture upon activation with thrombin or after laser-induced stimulation. PDI is localized in different cellular compartments in activated and quiescent endothelial cells, and is redistributed to the plasma membrane after cell activation. In vivo studies using intravital microscopy show that PDI appears rapidly after laser-induced vessel wall injury, before the appearance of the platelet thrombus. If platelet thrombus formation is inhibited by the infusion of eptifibatide into the circulation, PDI is detected after vessel wall injury, and fibrin deposition is normal. Treatment of mice with a function blocking anti-PDI antibody completely inhibits fibrin generation in eptifibatide-treated mice. These results indicate that, although both platelets and endothelial cells secrete PDI after laser-induced injury, PDI from endothelial cells is required for fibrin generation in vivo.


Asunto(s)
Plaquetas/metabolismo , Células Endoteliales/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/metabolismo , Animales , Línea Celular , Citosol/ultraestructura , Células Endoteliales/citología , Células Endoteliales/ultraestructura , Endotelio/metabolismo , Fibrina/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Disulfuro Isomerasas/análisis
19.
Clin Cancer Res ; 15(22): 6830-40, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19861441

RESUMEN

PURPOSE: Despite the strong association between malignant disease and thromboembolic disorders, the molecular and cellular basis of this relationship remains uncertain. We evaluated the hypothesis that tumor-derived tissue factor-bearing microparticles in plasma contribute to cancer-associated thrombosis. EXPERIMENTAL DESIGN: We developed impedance-based flow cytometry to detect, quantitate, and size microparticles in platelet-poor plasma. We evaluated the number of tissue factor-bearing microparticles in a cohort of cancer patients of different histologies (N = 96) and conducted a case-control study of 30 cancer patients diagnosed with an acute venous thromboembolic event (VTE) compared with 60 cancer patients of similar age, stage, sex, and diagnosis without known VTE, as well as 22 patients with an idiopathic VTE. RESULTS: Tissue factor-bearing microparticles were detected in patients with advanced malignancy, including two thirds of patients with pancreatic carcinoma. Elevated levels of tissue factor-bearing microparticles were associated VTE in cancer patients (adjusted odds ratio, 3.72; 95% confidence interval, 1.18-11.76; P = 0.01). In cancer patients without VTE, a retrospective analysis revealed a 1-year cumulative incidence of VTE of 34.8% in patients with tissue factor-bearing microparticles versus 0% in those without detectable tissue factor-bearing microparticles (Gray test P = 0.002).The median number of tissue factor-bearing microparticles in the cancer VTE cohort (7.1 x 10(4) microparticles/microL) was significantly greater than both the idiopathic VTE and cancer-no VTE groups (P = 0.002 and P = 0.03, respectively). Pancreatectomy in three patients eliminated or nearly eliminated these microparticles which coexpressed the epithelial tumor antigen, MUC-1. CONCLUSION: We conclude that tumor-derived tissue factor-bearing microparticles are associated with VTE in cancer patients and may be central to the pathogenesis of cancer-associated thrombosis.


Asunto(s)
Citometría de Flujo/métodos , Neoplasias/complicaciones , Neoplasias/patología , Tromboplastina/metabolismo , Tromboembolia Venosa/complicaciones , Tromboembolia Venosa/patología , Anciano , Biomarcadores de Tumor/metabolismo , Estudios de Casos y Controles , Técnicas de Cultivo de Célula/métodos , Separación Celular , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/cirugía , Factores de Riesgo , Trombosis , Tromboembolia Venosa/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...