Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 142(13): 1156-1166, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37506337

RESUMEN

von Willebrand factor (VWF) mediates primary hemostasis and thrombosis in response to hydrodynamic forces. We previously showed that high shear promoted self-association of VWF into hyperadhesive strands, which can be attenuated by high-density lipoprotein (HDL) and apolipoprotein A-I. In this study, we show that low-density lipoprotein (LDL) binds VWF under shear and enhances self-association. Vortexing VWF in tubes resulted in its loss from the solution and deposition onto tube surfaces, which was prevented by HDL. At a stabilizing HDL concentration of 1.2 mg/mL, increasing concentrations of LDL progressively increased VWF loss, the effect correlating with the LDL-to-HDL ratio and not the absolute concentration of the lipoproteins. Similarly, HDL diminished deposition of VWF in a post-in-channel microfluidic device, whereas LDL increased both the rate and extent of strand deposition, with both purified VWF and plasma. Hypercholesterolemic human plasma also displayed accelerated VWF accumulation in the microfluidic device. The initial rate of accumulation correlated linearly with the LDL-to-HDL ratio. In Adamts13-/- and Adamts13-/-LDLR-/- mice, high LDL levels enhanced VWF and platelet adhesion to the myocardial microvasculature, reducing cardiac perfusion, impairing systolic function, and producing early signs of cardiomyopathy. In wild-type mice, high plasma LDL concentrations also increased the size and persistence of VWF-platelet thrombi in ionophore-treated mesenteric microvessels, exceeding the accumulation seen in similarly treated ADAMTS13-deficient mice that did not receive LDL infusion. We propose that targeting the interaction of VWF with itself and with LDL may improve the course of thrombotic microangiopathies, atherosclerosis, and other disorders with defective microvascular circulation.


Asunto(s)
Trombosis , Factor de von Willebrand , Ratones , Humanos , Animales , Factor de von Willebrand/metabolismo , Lipoproteínas LDL , Trombosis/metabolismo , Hemostasis , Adhesividad Plaquetaria , Proteína ADAMTS13
2.
J Transl Med ; 22(1): 412, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693516

RESUMEN

BACKGROUND: Thromboinflammation involving platelet adhesion to endothelial surface-associated von Willebrand factor (VWF) has been implicated in the accelerated progression of non-culprit plaques after MI. The aim of this study was to use arterial endothelial molecular imaging to mechanistically evaluate endothelial-associated VWF as a therapeutic target for reducing remote plaque activation after myocardial infarction (MI). METHODS: Hyperlipidemic mice deficient for the low-density lipoprotein receptor and Apobec-1 underwent closed-chest MI and were treated chronically with either: (i) recombinant ADAMTS13 which is responsible for proteolytic removal of VWF from the endothelial surface, (ii) N-acetylcysteine (NAC) which removes VWF by disulfide bond reduction, (iii) function-blocking anti-factor XI (FXI) antibody, or (iv) no therapy. Non-ischemic controls were also studied. At day 3 and 21, ultrasound molecular imaging was performed with probes targeted to endothelial-associated VWF A1-domain, platelet GPIbα, P-selectin and vascular cell adhesion molecule-1 (VCAM-1) at lesion-prone sites of the aorta. Histology was performed at day 21. RESULTS: Aortic signal for P-selectin, VCAM-1, VWF, and platelet-GPIbα were all increased several-fold (p < 0.01) in post-MI mice versus sham-treated animals at day 3 and 21. Treatment with NAC and ADAMTS13 significantly attenuated the post-MI increase for all four molecular targets by > 50% (p < 0.05 vs. non-treated at day 3 and 21). On aortic root histology, mice undergoing MI versus controls had 2-4 fold greater plaque size and macrophage content (p < 0.05), approximately 20-fold greater platelet adhesion (p < 0.05), and increased staining for markers of platelet transforming growth factor-ß1 signaling. Accelerated plaque growth and inflammatory activation was almost entirely prevented by ADAMTS13 and NAC. Inhibition of FXI had no significant effect on molecular imaging signal or plaque morphology. CONCLUSIONS: Plaque inflammatory activation in remote arteries after MI is strongly influenced by VWF-mediated platelet adhesion to the endothelium. These findings support investigation into new secondary preventive therapies for reducing non-culprit artery events after MI.


Asunto(s)
Proteína ADAMTS13 , Infarto del Miocardio , Factor de von Willebrand , Animales , Factor de von Willebrand/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/complicaciones , Proteína ADAMTS13/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Ratones , Placa Aterosclerótica/patología , Selectina-P/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Masculino , Imagen Molecular , Aorta/patología , Aorta/efectos de los fármacos , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Ratones Endogámicos C57BL
3.
Blood ; 135(15): 1270-1280, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32077913

RESUMEN

Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder mediated by complexes between platelet factor 4 (PF4) and heparin or other polyanions, but the risk of thrombosis extends beyond exposure to heparin implicating other PF4 partners. We recently reported that peri-thrombus endothelium is targeted by HIT antibodies, but the binding site(s) has not been identified. We now show that PF4 binds at multiple discrete sites along the surface of extended strings of von Willebrand factor (VWF) released from the endothelium following photochemical injury in an endothelialized microfluidic system under flow. The HIT-like monoclonal antibody KKO and HIT patient antibodies recognize PF4-VWF complexes, promoting platelet adhesion and enlargement of thrombi within the microfluidic channels. Platelet adhesion to the PF4-VWF-HIT antibody complexes is inhibited by antibodies that block FcγRIIA or the glycoprotein Ib-IX complex on platelets. Disruption of PF4-VWF-HIT antibody complexes by drugs that prevent or block VWF oligomerization attenuate thrombus formation in a murine model of HIT. Together, these studies demonstrate assembly of HIT immune complexes along VWF strings released by injured endothelium that might propagate the risk of thrombosis in HIT. Disruption of PF4-VWF complex formation may provide a new therapeutic approach to HIT.


Asunto(s)
Anticuerpos/inmunología , Anticoagulantes/efectos adversos , Heparina/efectos adversos , Factor Plaquetario 4/inmunología , Trombocitopenia/inducido químicamente , Trombosis/etiología , Factor de von Willebrand/inmunología , Animales , Anticoagulantes/inmunología , Heparina/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Adhesividad Plaquetaria , Trombocitopenia/complicaciones , Trombocitopenia/inmunología , Trombocitopenia/patología , Trombosis/inmunología , Trombosis/patología
4.
Blood ; 132(2): 141-147, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29866815

RESUMEN

Increasing evidence indicates that inflammation can cause thrombosis by a von Willebrand factor (VWF)-mediated mechanism that includes endothelial activation, secretion of VWF, assembly of hyperadhesive VWF strings and fibers, cleavage by ADAMTS13, and adhesion and deposition of VWF-platelet thrombi in the vasculature. This mechanism appears to contribute to thrombosis not only in small vessels, but also in large vessels. Inflammation and VWF contribute to atherogenesis and may contribute to arterial and venous thrombosis as well as stroke. Elucidation of the mechanism will hopefully identify new targets and suggest new approaches for prevention and intervention.


Asunto(s)
Proteína ADAMTS13/genética , Proteína ADAMTS13/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Plaquetas/metabolismo , Regulación de la Expresión Génica , Humanos , Oxidación-Reducción , Unión Proteica , Biosíntesis de Proteínas , Proteolisis , Transcripción Genética
5.
Blood ; 130(21): 2295-2306, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-28924019

RESUMEN

Lymphodepletion chemotherapy followed by infusion of CD19-specific chimeric antigen receptor-modified (CAR) T cells has produced impressive antitumor responses in patients with refractory CD19+ B-cell malignancies but is often associated with cytokine release syndrome (CRS). Our understanding of CRS continues to evolve, and identification of the kinetics of CRS and predictive clinical and laboratory biomarkers of severity are needed to evaluate strategies to mitigate toxicity. We report the clinical presentation of and identify biomarkers of severe CRS in 133 adult patients who received CD19 CAR T cells. CRS developed in 70% of patients, including 62.5% with grade 1 to 3 CRS (grade 1, 26%; grade 2, 32%; grade 3, 4.5%), 3.8% with grade 4, and 3.8% with grade 5. A majority of cases of grade ≥4 CRS occurred during CAR T-cell dose finding. Multivariable analysis of baseline characteristics identified high marrow tumor burden, lymphodepletion using cyclophosphamide and fludarabine, higher CAR T-cell dose, thrombocytopenia before lymphodepletion, and manufacturing of CAR T cells without selection of CD8+ central memory T cells as independent predictors of CRS. Severe CRS was characterized by hemodynamic instability, capillary leak, and consumptive coagulopathy. Angiopoietin-2 and von Willebrand factor, which are biomarkers of endothelial activation, were increased during severe CRS and also before lymphodepletion in patients who subsequently developed CRS. We describe a classification-tree algorithm to guide studies of early intervention after CAR T-cell infusion for patients at high risk of severe CRS. These data provide a framework for early intervention studies to facilitate safer application of effective CD19 CAR T-cell therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Citocinas/metabolismo , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Adulto , Anciano , Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/fisiopatología , Trastornos de la Coagulación Sanguínea/terapia , Células Endoteliales/patología , Femenino , Fiebre/etiología , Hematopoyesis , Hemodinámica , Humanos , Cinética , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Síndromes de Neurotoxicidad/complicaciones , Síndromes de Neurotoxicidad/fisiopatología , Síndromes de Neurotoxicidad/terapia , Factores de Riesgo , Síndrome , Resultado del Tratamiento , Adulto Joven
6.
Int J Med Sci ; 16(2): 276-284, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30745808

RESUMEN

Background: Endothelial activation caused by HIV-1 infection leads to release of von Willebrand factor (VWF), which enters the circulation or attaches to vessel walls and self-assembles into strings and fibers, enabling platelet adhesion; this adhesive activity is regulated by the VWF-cleaving protease ADAMTS13. Our objective was to assess VWF adhesive activity and ADAMTS13 protease activity in HIV-1 infection. Methods: We measured levels of VWF antigen, VWF activation factor (a measure of adhesive activity), ADAMTS13 antigen, ADAMTS13 activity, and apolipoprotein A1 (which interferes with VWF self-association) in serum samples from HIV-1-infected men whose infections were acute (n=10), chronic untreated (n=10), or chronic treated (n=10), compared to uninfected controls (n=10). Means across groups were compared using analysis of variance with contrasts, and Pearson correlations were calculated. Results: Plasma viral load was positively correlated with VWF adhesive activity, which was elevated in acute relative to chronic treated HIV-1 infection. ADAMTS13 antigen and activity were both positively correlated with plasma viral load, and ADAMTS13 activity was significantly higher in men with acute HIV infection than in uninfected controls, and in both acute and chronic untreated HIV infection relative to chronic treated infection. Conclusion: These findings suggest that even in the setting of increased ADAMTS13 protease activity, VWF in HIV-1 infection is hyperadhesive, which may favor development of microvascular and arterial thromboses and thereby contribute to increased cardiovascular risk in HIV-1-infected individuals.


Asunto(s)
Proteína ADAMTS13/metabolismo , Infecciones por VIH/sangre , Factor de von Willebrand/metabolismo , Proteína ADAMTS13/inmunología , Adulto , Apolipoproteína A-I/sangre , Biomarcadores/sangre , Estudios de Casos y Controles , VIH-1 , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Factor de von Willebrand/inmunología
7.
Blood ; 127(5): 637-45, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26552698

RESUMEN

The ability of von Willebrand factor (VWF) to initiate platelet adhesion depends on the number of monomers in individual VWF multimers and on the self-association of individual VWF multimers into larger structures. VWF self-association is accelerated by shear stress. We observed that VWF self-association occurs during adsorption of VWF onto surfaces, assembly of secreted VWF into hyperadhesive VWF strings on the endothelial surface, and incorporation of fluid-phase VWF into VWF fibers. VWF adsorption under static conditions increased with increased VWF purity and was prevented by a component of plasma. We identified that component as high-density lipoprotein (HDL) and its major apolipoprotein ApoA-I. HDL and ApoA-I also prevented VWF on the endothelium from self-associating into longer strands and inhibited the attachment of fluid-phase VWF onto vessel wall strands. Platelet adhesion to VWF fibers was reduced in proportion to the reduction in self-associated VWF. In a mouse model of thrombotic microangiopathy, HDL also largely prevented the thrombocytopenia induced by injection of high doses of human VWF. Finally, a potential role for ApoA-I in microvascular occlusion associated with thrombotic thrombocytopenic purpura and sepsis was revealed by the inverse relationship between the concentration of ApoA-I and that of hyperadhesive VWF. These results suggest that interference with VWF self-association would be a new approach to treating thrombotic disorders.


Asunto(s)
Apolipoproteína A-I/metabolismo , Lipoproteínas HDL/metabolismo , Adhesividad Plaquetaria , Trombosis/metabolismo , Factor de von Willebrand/metabolismo , Animales , Apolipoproteína A-I/uso terapéutico , Plaquetas/citología , Plaquetas/metabolismo , Humanos , Lipoproteínas HDL/uso terapéutico , Ratones Endogámicos C57BL , Multimerización de Proteína , Trombocitopenia/prevención & control , Factor de von Willebrand/química
8.
Blood ; 125(6): 1034-7, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25395424

RESUMEN

Several complement proteins interact with hemostatic factors. We discovered that von Willebrand factor (VWF) acts as a cofactor for factor I-mediated cleavage of complement C3b, thereby shutting down complement activation. The complement regulatory function of VWF multimers depends on their size. Smaller VWF multimers enhance cleavage of C3b but large and ultra-large VWF (ULVWF) multimers have no effect on C3b cleavage and permit default complement activation. We conclude that normal plasma VWF multimers prevent complement activation and steer the complement pathway toward generation of inactivated C3b (iC3b). ULVWF multimers, as are present in patients with thrombotic microangiopathy, lack an inhibitory effect on complement and permit complement activation.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Fibrinógeno/inmunología , Factor de von Willebrand/inmunología , Proteínas del Sistema Complemento/metabolismo , Fibrinógeno/metabolismo , Humanos , Multimerización de Proteína , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
9.
Blood ; 126(2): 262-9, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26019279

RESUMEN

Von Willebrand disease (VWD) is an inherited bleeding disorder characterized by incomplete penetrance and variable expressivity. We evaluated a 24-member pedigree with VWD type 2 caused by a T>G mutation at position 3911 that predicts a methionine to arginine (M1304R) change in the platelet-binding A1 domain of von Willebrand factor (VWF). This mutation manifests as an autosomal-dominant trait, with clinical and biochemical phenotypic variability among affected individuals, including differences in bleeding tendency and VWF quantity, activity, and multimer pattern. Sequencing of all VWF coding regions in 3 affected individuals did not identify additional mutations. When expressed in heterologous cells, M1304R was secreted in lower quantities, failed to drive formation of storage granules, and was defective in multimerization and platelet binding. When cotransfected in equal quantities with the wild-type complementary DNA, the mutant complementary DNA depressed VWF secretion, although multimerization was only mildly affected. A llama nanobody (AU/VWFa-11) that detects the mutant A1 domain demonstrated highly variable binding to VWF from different affected members, indicating that the VWF contained different percentages of mutant monomers in different individuals. Thus, the observed variability in VWD phenotypes could in part be determined by the extent of mutant monomer incorporation in the final multimer structure of plasma VWF.


Asunto(s)
Familia , Proteínas Mutantes/metabolismo , Multimerización de Proteína , Enfermedades de von Willebrand , Factor de von Willebrand/metabolismo , Cristalografía por Rayos X , Femenino , Células HEK293 , Humanos , Masculino , Simulación del Acoplamiento Molecular , Mutación Missense , Linaje , Fenotipo , Unión Proteica , Multimerización de Proteína/fisiología , Estructura Terciaria de Proteína , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/genética , Enfermedades de von Willebrand/metabolismo , Factor de von Willebrand/química , Factor de von Willebrand/genética
10.
J Biol Chem ; 290(3): 1422-31, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25422322

RESUMEN

ADAMTS13 is a plasma metalloproteinase that cleaves large multimeric forms of von Willebrand factor (VWF) to smaller, less adhesive forms. ADAMTS13 activity is reduced in systemic inflammatory syndromes, but the cause is unknown. Here, we examined whether neutrophil-derived oxidants can regulate ADAMTS13 activity. We exposed ADAMTS13 to hypochlorous acid (HOCl), produced by a myeloperoxidase-H2O2-Cl(-) system, and determined its residual proteolytic activity using both a VWF A2 peptide substrate and multimeric plasma VWF. Treatment with 25 nm myeloperoxidase plus 50 µm H2O2 reduced ADAMTS13 activity by >85%. Using mass spectrometry, we demonstrated that Met(249), Met(331), and Met(496) in important functional domains of ADAMTS13 were oxidized to methionine sulfoxide in an HOCl concentration-dependent manner. The loss of enzyme activity correlated with the extent of oxidation of these residues. These Met residues were also oxidized in ADAMTS13 exposed to activated human neutrophils, accompanied by reduced enzyme activity. ADAMTS13 treated with either neutrophil elastase or plasmin was inhibited to a lesser extent, especially in the presence of plasma. These observations suggest that oxidation could be an important mechanism for ADAMTS13 inactivation during inflammation and contribute to the prothrombotic tendency associated with inflammation.


Asunto(s)
Proteínas ADAM/metabolismo , Regulación Enzimológica de la Expresión Génica , Ácido Hipocloroso/química , Inflamación/metabolismo , Neutrófilos/metabolismo , Oxidantes/química , Proteína ADAMTS13 , Cromatografía Liquida , Fibrinolisina/metabolismo , Humanos , Peróxido de Hidrógeno/química , Elastasa de Leucocito/química , Espectrometría de Masas , Metionina/química , Neutrófilos/enzimología , Oxígeno/química , Peroxidasa/metabolismo , Estructura Terciaria de Proteína , Trombosis/metabolismo , Factor de von Willebrand/metabolismo
11.
Blood ; 133(17): 1800-1801, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-31023744
12.
Malar J ; 15: 56, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26830467

RESUMEN

BACKGROUND: Malaria is a major cause of morbidity and mortality in sub-Saharan Africa, and poor outcomes have been associated with endothelial activation. In this study, biomarkers of endothelial activation, haemostasis, and thrombosis were measured in Ugandan children with severe malaria who participated in a clinical trial, in order to investigate associations between these processes. METHODS: Serum and plasma were collected from participants at baseline (day 1), and on days 2, 3, 4, and 14. Von Willebrand factor (VWF) antigen was measured in stored plasma samples from all trial participants, and its association with mortality and changes over time were analysed. VWF multimer patterns were evaluated in baseline serum samples by gel electrophoresis followed by Western blotting. Levels of angiopoietins 1 and 2, VWF antigen, total active VWF, ADAMTS13, platelet counts, apolipoprotein A1, and syndecan-1 were measured in stored serum samples from 12 survivors at baseline and day 4. RESULTS: VWF antigen levels were associated with mortality, and decreased over time in survivors. Baseline VWF antigen and total active VWF levels were elevated, and very large multimers were present in the baseline serum of several patients. Higher platelet counts were associated with higher angiopoietin-1 and apolipoprotein A1 levels, while lower platelet counts were associated with higher syndecan-1, a marker of endothelial damage. Higher angiopoietin-2 to angiopoietin-1 ratio and higher syndecan-1 levels were correlated with lower apolipoprotein A1 levels. There were no correlations between total active VWF, VWF antigen, or ADAMTS13 levels and the other biomarkers at baseline. Changes in biomarker levels between baseline and day 4 were not correlated. CONCLUSIONS: These results confirm that severe malaria is associated with endothelial activation, and suggest that endothelial activation contributes to microvascular thrombosis and endothelial damage.


Asunto(s)
Biomarcadores/sangre , Hemostasis/fisiología , Malaria/sangre , Malaria/fisiopatología , Trombosis/sangre , Proteína ADAMTS13/sangre , Angiopoyetina 1/sangre , Angiopoyetina 2/sangre , Western Blotting , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Sindecano-1/sangre , Trombosis/fisiopatología , Uganda/epidemiología , Factor de von Willebrand/metabolismo
13.
bioRxiv ; 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38370737

RESUMEN

Protein S (PS), the critical plasma cofactor for the anticoagulants tissue factor (TF) pathway inhibitor (TFPI) and activated protein C (APC), circulates in two functionally distinct pools: free (anticoagulant) or bound to complement component 4b-binding protein (C4BP) (anti-inflammatory). Acquired free PS deficiency is detected in several viral infections, but its cause is unclear. Here, we identified a shear-dependent interaction between PS and von Willebrand Factor (VWF) by mass spectrometry. Consistently, plasma PS and VWF comigrated in both native and agarose gel electrophoresis. The PS/VWF interaction was blocked by TFPI but not APC, suggesting an interaction with the C-terminal sex hormone binding globulin (SHBG) region of PS. Microfluidic systems, mimicking arterial laminar flow or disrupted turbulent flow, demonstrated that PS stably binds VWF as VWF unfolds under turbulent flow. PS/VWF complexes also localized to platelet thrombi under laminar arterial flow. In thrombin generation-based assays, shearing plasma decreased PS activity, an effect not seen in the absence of VWF. Finally, free PS deficiency in COVID-19 patients, measured using an antibody that binds near the C4BP binding site in SHBG, correlated with changes in VWF, but not C4BP, and with thrombin generation. Our data suggest that PS binds to a shear-exposed site on VWF, thus sequestering free PS and decreasing its anticoagulant activity, which would account for the increased thrombin generation potential. As many viral infections present with free PS deficiency, elevated circulating VWF, and increased vascular shear, we propose that the PS/VWF interaction reported here is a likely contributor to virus-associated thrombotic risk.

14.
Blood ; 118(19): 5283-91, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21917758

RESUMEN

VWF is required for platelet adhesion to sites of vessel injury, a process vital for both hemostasis and thrombosis. Enhanced VWF secretion and oxidative stress are both hallmarks of inflammation. We recently showed that the neutrophil oxidant hypochlorous acid (HOCl) inhibits VWF proteolysis by ADAMTS13 by oxidizing VWF methionine 1606 (M1606) in the A2 domain. M1606 was readily oxidized in a substrate peptide, but required urea in multimeric plasma VWF. In the present study, we examined whether shear stress enhances VWF oxidation. With an HOCl-generating system containing myeloperoxidase (MPO) and H(2)O(2), we found that shear stress accelerated M1606 oxidation, with 56% becoming oxidized within 1 hour. Seven other methionine residues in the VWF A1A2A3 region (containing the sites for platelet and collagen binding and ADAMTS13 cleavage) were variably oxidized, one completely. Oxidized methionines accumulated preferentially in the largest VWF multimers. HOCl-oxidized VWF was hyperfunctional, agglutinating platelets at ristocetin concentrations that induced minimal agglutination using unoxidized VWF and binding more of the nanobody AU/VWFa-11, which detects a gain-of-function conformation of the A1 domain. These findings suggest that neutrophil oxidants will both render newly secreted VWF uncleavable and alter the largest plasma VWF forms such that they become hyperfunctional and resistant to proteolysis by ADAMTS13.


Asunto(s)
Factor de von Willebrand/química , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Sitios de Unión , Plaquetas/metabolismo , Humanos , Ácido Hipocloroso , Técnicas In Vitro , Metionina/química , Modelos Moleculares , Oxidación-Reducción , Multimerización de Proteína , Desplegamiento Proteico , Proteolisis , Resistencia al Corte , Estrés Mecánico , Factor de von Willebrand/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 32(11): 2625-30, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22922961

RESUMEN

OBJECTIVE: Platelet-bound von Willebrand factor (VWF) was recently demonstrated to be a better substrate for ADAMTS13, suggesting that 1 conformational change exposes both the glycoprotein Ibα binding site in the A1 domain and the ADAMTS13 cleavage site in the A2 domain. Because ristocetin induces VWF to bind glycoprotein Ibα in the absence of shear stress, we evaluated whether it could also enhance ADAMTS13 proteolysis of VWF. METHODS AND RESULTS: We used several VWF sources: plasma, purified plasma VWF, recombinant VWF fragments encompassing A1A2A3, A1A2, and 2 A2 domains, 1 containing a ristocetin-binding site (Asp1459-His1472) and the other lacking it. Ristocetin accelerated ADAMTS13 cleavage of multimeric VWF and of each of the recombinant VWF fragments except for the A2 domain lacking the ristocetin-binding site. We also examined the effect of ristocetin on the conformation of the A2 domain by assessing its effect on the susceptibility of Met1606 at the ADAMTS13 cleavage site to be oxidized by hypochlorous acid. Ristocetin markedly enhanced oxidation of Met1606 and Met1521 of the A2 domain. CONCLUSIONS: These data indicate that exposure of the sites for glycoprotein Ibα and ADAMTS13 are coupled, explaining why platelet-bound VWF is a better ADAMTS13 substrate and why enhanced proteolysis is often observed in type 2B von Willebrand disease.


Asunto(s)
Proteínas ADAM/metabolismo , Glicoproteínas de Membrana/metabolismo , Ristocetina/metabolismo , Factor de von Willebrand/metabolismo , Proteína ADAMTS13 , Sitios de Unión , Humanos , Ácido Hipocloroso/química , Oxidación-Reducción , Complejo GPIb-IX de Glicoproteína Plaquetaria , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Desplegamiento Proteico , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Factores de Tiempo , Factor de von Willebrand/química
16.
Biophys J ; 103(11): 2399-407, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23283239

RESUMEN

Using a combination of structural and mechanical characterization, we examine the effect of fibrinogen oxidation on the formation of fibrin clots. We find that treatment with hypochlorous acid preferentially oxidizes specific methionine residues on the α, ß, and γ chains of fibrinogen. Oxidation is associated with the formation of a dense network of thin fibers after activation by thrombin. Additionally, both the linear and nonlinear mechanical properties of oxidized fibrin gels are found to be altered with oxidation. Finally, the structural modifications induced by oxidation are associated with delayed fibrin lysis via plasminogen and tissue plasminogen activator. Based on these results, we speculate that methionine oxidation of specific residues may be related to hindered lateral aggregation of protofibrils in fibrin gels.


Asunto(s)
Coagulación Sanguínea , Fibrina/química , Fibrinógeno/química , Metionina/química , Plasminógeno/química , Activador de Tejido Plasminógeno/química , Sitios de Unión , Humanos , Oxidación-Reducción , Unión Proteica
18.
Blood ; 115(3): 706-12, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19812385

RESUMEN

Elevated plasma von Willebrand factor (VWF) and low ADAMTS13 activity have been reported in several inflammatory states, including sepsis and acute respiratory distress syndrome. One hallmark of inflammation is neutrophil activation and production of reactive oxygen species, including superoxide radical, hydrogen peroxide, and hypochlorous acid (HOCl). HOCl is produced from hydrogen peroxide and chloride ions through the action of myeloperoxidase. HOCl can oxidize methionine to methionine sulfoxide and tyrosine to chlorotyrosine. This is of interest because the ADAMTS13 cleavage site in VWF, the Tyr(1605)-Met(1606) peptide bond, contains both oxidation-prone residues. We hypothesized that HOCl would oxidize either or both of these residues and possibly inhibit ADAMTS13-mediated cleavage. We therefore treated ADAMTS13 substrates with HOCl and examined their oxidative modification by mass spectrometry. Met(1606) was oxidized to the sulfoxide in a concentration-dependent manner, with complete oxidation at 75muM HOCl, whereas only a miniscule percentage of Tyr(1605) was converted to chlorotyrosine. The oxidized substrates were cleaved much more slowly by ADAMTS13 than the nonoxidized substrates. A similar result was obtained with multimeric VWF. Taken together, these findings indicate that reactive oxygen species released by activated neutrophils have a prothrombotic effect, mediated in part by inhibition of VWF cleavage by ADAMTS13.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Neutrófilos/metabolismo , Oxidantes/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Factor de von Willebrand/metabolismo , Proteínas ADAM/química , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Secuencia de Aminoácidos , Dominio Catalítico , Células Cultivadas , Humanos , Peróxido de Hidrógeno/farmacología , Ácido Hipocloroso/farmacología , Metionina/metabolismo , Metionina/fisiología , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Oxidantes/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Multimerización de Proteína/fisiología , Especies Reactivas de Oxígeno/farmacología , Tirosina/metabolismo , Tirosina/fisiología , Factor de von Willebrand/química
19.
JACC Basic Transl Sci ; 7(7): 642-655, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35958695

RESUMEN

We hypothesized that excess endothelial-associated von Willebrand factor (vWF) and secondary platelet adhesion contribute to aortic valve stenosis (AS). We studied hyperlipidemic mice lacking ADAMTS13 (LDLR -/- AD13 -/- ), which cleaves endothelial-associated vWF multimers. On echocardiography and molecular imaging, LDLR -/- AD13 -/- compared with control strains had increased aortic endothelial vWF and platelet adhesion and developed hemodynamically significant AS, arterial stiffening, high valvulo-aortic impedance, and secondary load-dependent reduction in LV systolic function. Histology revealed leaflet thickening and calcification with valve interstitial cell myofibroblastic and osteogenic transformation, and evidence for TGFß1 pathway activation. We conclude that valve leaflet endothelial vWF-platelet interactions promote AS through juxtacrine platelet signaling.

20.
J Neurotrauma ; 39(11-12): 879-890, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35316073

RESUMEN

Traumatic brain injury (TBI) impairs cerebrovascular autoregulation and reduces cerebral blood flow (CBF), leading to ischemic secondary injuries. We have shown that injured brains release brain-derived extracellular vesicles (BDEVs) into circulation, where they cause a systemic hypercoagulable state that rapidly turns into consumptive coagulopathy. The BDEVs induce endothelial injury and permeability, leading to the hypothesis that they contribute to TBI-induced cerebrovascular dysregulation. In a study designed to test this hypothesis, we detected circulating BDEVs in C57BL/6J mice subjected to severe TBI, reaching peak levels of 3 × 104/µL at 3 h post-injury (71.2 ± 21.5% of total annexin V-binding EVs). We further showed in an adaptive transfer model that 41.7 ± 5.8% of non-injured mice died within 6 h after being infused with 3 × 104/µL of BDEVs. The BDEVs transmigrated through the vessel walls, induced rapid vasoconstriction by inducing calcium influx in vascular smooth muscle cells, and reduced CBF by 93.8 ± 5.6% within 30 min after infusion. The CBF suppression was persistent in mice that eventually died, but it recovered quickly in surviving mice. It was prevented by the calcium channel blocker nimodipine. When being separated, neither protein nor phospholipid components from the lethal number of BDEVs induced vasoconstriction, reduced CBF, and caused death. These results demonstrate a novel vasoconstrictive activity of BDEVs that depends on the structure of BDEVs and contributes to TBI-induced disseminated cerebral ischemia and sudden death.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Vesículas Extracelulares , Animales , Encéfalo , Circulación Cerebrovascular/fisiología , Vesículas Extracelulares/metabolismo , Ratones , Ratones Endogámicos C57BL , Vasoconstricción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA