Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3297, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740748

RESUMEN

Despite abundant evidence demonstrating that platelets foster metastasis, anti-platelet agents have low therapeutic potential due to the risk of hemorrhages. In addition, whether platelets can regulate metastasis at the late stages of the disease remains unknown. In this study, we subject syngeneic models of metastasis to various thrombocytopenic regimes to show that platelets provide a biphasic contribution to metastasis. While potent intravascular binding of platelets to tumor cells efficiently promotes metastasis, platelets further support the outgrowth of established metastases via immune suppression. Genetic depletion and pharmacological targeting of the glycoprotein VI (GPVI) platelet-specific receptor in humanized mouse models efficiently reduce the growth of established metastases, independently of active platelet binding to tumor cells in the bloodstream. Our study demonstrates therapeutic efficacy when targeting animals bearing growing metastases. It further identifies GPVI as a molecular target whose inhibition can impair metastasis without inducing collateral hemostatic perturbations.


Asunto(s)
Plaquetas , Metástasis de la Neoplasia , Glicoproteínas de Membrana Plaquetaria , Animales , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Humanos , Ratones , Glicoproteínas de Membrana Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/genética , Línea Celular Tumoral , Femenino , Ratones Endogámicos C57BL
2.
Thromb Res ; 221: 137-148, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36376109

RESUMEN

Severe COVID-19 has been associated with a high rate of thrombotic events but also of bleeding events, particularly when the level of prophylactic anticoagulation was increased. Data on the contribution of platelets to these thrombotic events are discordant between reports, while the involvement of platelets in bleeding events has never been investigated. The objective of the present study was to assess platelet function during the first week of ICU hospitalization in patients with severe COVID-19 pneumonia. A total of 35 patients were prospectively included and blood samples were drawn on day (D) 0, D2 and D7. COVID-19 pneumonia was severe with a median PaO2/FiO2 ratio of 91 [68-119] on D0. Platelets from these patients showed evidence of pre-activation and exhaustion with a significant reduction in the surface expression of GPVI, GPIb and GPIIbIIIa, together with a decrease in serotonin content. Platelets from patients with severe COVID-19 were hyporesponsive with a reduced maximal aggregation response to several platelet agonists and decreased adhesion to immobilized fibrinogen. Aggregation of washed platelets and plasma substitution experiments indicated that a plasma factor was at least partially responsible for this hyporeactivity of platelets. Blood flow experiments showed that severe COVID-19 platelets formed smaller, less stable aggregates on a collagen-coated surface, which could explain why some patients develop bleeding events. These findings should prompt us to carefully evaluate the risks and benefits of high-dose prophylactic anticoagulation, and to decrease the level of anticoagulation once the initial phase of the disease has resolved. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT04359992.


Asunto(s)
COVID-19 , Trombosis , Humanos , Anticoagulantes/metabolismo , Coagulación Sanguínea , Plaquetas/metabolismo , COVID-19/complicaciones , Hemorragia/metabolismo , Agregación Plaquetaria , Estudios Prospectivos
3.
Thromb Haemost ; 122(5): 767-776, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34598304

RESUMEN

OBJECTIVE: Integrins are key regulators of various platelet functions. The pathophysiological importance of most platelet integrins has been investigated, with the exception of α5ß1, a receptor for fibronectin. The aim of this study was to characterize the role of α5ß1 in megakaryopoiesis, platelet function, and to determine its importance in hemostasis and arterial thrombosis. APPROACH AND RESULTS: We generated a mouse strain deficient for integrin α5ß1 on megakaryocytes and platelets (PF4Cre-α5-/-). PF4Cre-α5-/- mice were viable, fertile, and presented no apparent signs of abnormality. Megakaryopoiesis appears unaltered as evidence by a normal megakaryocyte morphology and development, which is in agreement with a normal platelet count. Expression of the main platelet receptors and the response of PF4Cre-α5-/- platelets to a series of agonists were all completely normal. Adhesion and aggregation of PF4Cre-α5-/- platelets under shear flow on fibrinogen, laminin, or von Willebrand factor were unimpaired. In contrast, PF4Cre-α5-/- platelets displayed a marked decrease in adhesion, activation, and aggregation on fibrillar cellular fibronectin and collagen. PF4Cre-α5-/- mice presented no defect in a tail-bleeding time assay and no increase in inflammatory bleeding in a reverse passive Arthus model and a lipopolysaccharide pulmonary inflammation model. Finally, no defects were observed in three distinct experimental models of arterial thrombosis based on ferric chloride-induced injury of the carotid artery, mechanical injury of the abdominal aorta, or laser-induced injury of mesenteric vessels. CONCLUSION: In summary, this study shows that platelet integrin α5ß1 is a key receptor for fibrillar cellular fibronectin but is dispensable in hemostasis and arterial thrombosis.


Asunto(s)
Adhesividad Plaquetaria , Trombosis , Animales , Plaquetas/metabolismo , Fibronectinas/metabolismo , Hemostasis , Humanos , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrinas/metabolismo , Ratones , Trombosis/metabolismo
4.
Nature ; 581(7806): 58-62, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32376963

RESUMEN

When miniaturizing fluidic circuitry, the solid walls of the fluid channels become increasingly important1 because they limit the flow rates achievable for a given pressure drop, and they are prone to fouling2. Approaches for reducing the wall interactions include hydrophobic coatings3, liquid-infused porous surfaces4-6, nanoparticle surfactant jamming7, changes to surface electronic structure8, electrowetting9,10, surface tension pinning11,12 and use of atomically flat channels13. A better solution may be to avoid the solid walls altogether. Droplet microfluidics and sheath flow achieve this but require continuous flow of the central liquid and the surrounding liquid1,14. Here we demonstrate an approach in which aqueous liquid channels are surrounded by an immiscible magnetic liquid, both of which are stabilized by a quadrupolar magnetic field. This creates self-healing, non-clogging, anti-fouling and near-frictionless liquid-in-liquid fluidic channels. Manipulation of the field provides flow control, such as valving, splitting, merging and pumping. The latter is achieved by moving permanent magnets that have no physical contact with the liquid channel. We show that this magnetostaltic pumping method can be used to transport whole human blood with very little damage due to shear forces. Haemolysis (rupture of blood cells) is reduced by an order of magnitude compared with traditional peristaltic pumping, in which blood is mechanically squeezed through a plastic tube. Our liquid-in-liquid approach provides new ways to transport delicate liquids, particularly when scaling channels down to the micrometre scale, with no need for high pressures, and could also be used for microfluidic circuitry.

5.
Blood Adv ; 3(15): 2368-2380, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31399401

RESUMEN

The biogenesis of lysosome related organelles is defective in Hermansky-Pudlak syndrome (HPS), a disorder characterized by oculocutaneous albinism and platelet dense granule (DG) defects. The first animal model of HPS was the fawn-hooded rat, harboring a spontaneous mutation inactivating the small guanosine triphosphatase Rab38 This leads to coat color dilution associated with the absence of DGs and lung morphological defects. Another RAB38 mutant, the cht mouse, has normal DGs, which has raised controversy about the role of RAB38 in DG biogenesis. We show here that murine and human, but not rat, platelets also express the closely related RAB32. To elucidate the parts played by RAB32 and RAB38 in the biogenesis of DGs in vivo and their effects on platelet functions, we generated mice inactivated for Rab32, Rab38, and both genes. Single Rab38 inactivation mimicked cht mice, whereas single Rab32 inactivation had no effect in DGs, coat color, or lung morphology. By contrast, Rab32/38 double inactivation mimicked severe HPS, with strong coat and eye pigment dilution, some enlarged lung multilamellar bodies associated with a decrease in the number of DGs. These organelles were morphologically abnormal, decreased in number, and devoid of 5-hydroxytryptamine content. In line with the storage pool defect, platelet activation was affected, resulting in severely impaired thrombus growth and prolongation of the bleeding time. Overall, our study demonstrates the absence of impact of RAB38 or RAB32 single deficiency in platelet biogenesis and function resulting from full redundancy, and characterized a new mouse model mimicking HPS devoid of DG content.


Asunto(s)
Predisposición Genética a la Enfermedad , Síndrome de Hermanski-Pudlak/genética , Trombosis/genética , Proteínas de Unión al GTP rab/genética , Animales , Plaquetas/metabolismo , Plaquetas/ultraestructura , Modelos Animales de Enfermedad , Estudios de Asociación Genética/métodos , Síndrome de Hermanski-Pudlak/diagnóstico , Síndrome de Hermanski-Pudlak/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación , Fenotipo , Recuento de Plaquetas , Pruebas de Función Plaquetaria , Ratas , Trombosis/diagnóstico , Trombosis/metabolismo , Proteínas de Unión al GTP rab/metabolismo
6.
Matrix Biol ; 83: 26-47, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31288084

RESUMEN

Metastasis is a major cause of death in cancer patients. The extracellular matrix molecule tenascin-C is a known promoter of metastasis, however the underlying mechanisms are not well understood. To further analyze the impact of tenascin-C on cancer progression we generated MMTV-NeuNT mice that develop spontaneous mammary tumors, on a tenascin-C knockout background. We also developed a syngeneic orthotopic model in which tumor cells derived from a MMTV-NeuNT tumor. Tumor cells were transfected with control shRNA or with shRNA to knockdown tenascin-C expression and, were grafted into the mammary gland of immune competent, wildtype or tenascin-C knockout mice. We show that stromal-derived tenascin-C increases metastasis by reducing apoptosis and inducing the cellular plasticity of cancer cells located in pulmonary blood vessels invasions (BVI), before extravasation. We characterized BVI as organized structures of tightly packed aggregates of proliferating tumor cells with epithelial characteristics, surrounded by Fsp1+ cells, internally located platelets and, a luminal monolayer of endothelial cells. We found extracellular matrix, in particular, tenascin-C, between the stromal cells and the tumor cell cluster. In mice lacking stromal-derived tenascin-C, the organization of pulmonary BVI was significantly affected, revealing novel functions of host-derived tenascin-C in supporting the integrity of the endothelial cell coat, increasing platelet abundance, tumor cell survival, epithelial plasticity, thereby promoting overall lung metastasis. Many effects of tenascin-C observed in BVI including enhancement of cellular plasticity, survival and migration, could be explained by activation of TGF-ß signaling. Finally, in several human cancers, we also observed BVI to be surrounded by an endothelial monolayer and to express tenascin-C. Expression of tenascin-C is specific to BVI and is not observed in lymphatic vascular invasions frequent in breast cancer, which lack an endothelial lining. Given that BVI have prognostic significance for many tumor types, such as shorter cancer patient survival, increased metastasis, vessel occlusion, and organ failure, our data revealing a novel mechanism by which stromal tenascin-C promotes metastasis in human cancer, may have potential for diagnosis and therapy.


Asunto(s)
Vasos Sanguíneos/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Receptor ErbB-2/genética , Tenascina/genética , Animales , Vasos Sanguíneos/metabolismo , Línea Celular Tumoral , Femenino , Técnicas de Inactivación de Genes , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/genética , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Ratas , Transducción de Señal , Células del Estroma , Tenascina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
JCI Insight ; 1(14): e88245, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27699237

RESUMEN

Metastatic dissemination of cancer cells, which accounts for 90% of cancer mortality, is the ultimate hallmark of malignancy. Growing evidence suggests that blood platelets have a predominant role in tumor metastasis; however, the molecular mechanisms involved remain elusive. Here, we demonstrate that genetic deficiency of integrin α6ß1 on platelets markedly decreases experimental and spontaneous lung metastasis. In vitro and in vivo assays reveal that human and mouse platelet α6ß1 supports platelet adhesion to various types of cancer cells. Using a knockdown approach, we identified ADAM9 as the major counter receptor of α6ß1 on both human and mouse tumor cells. Static and flow-based adhesion assays of platelets binding to DC-9, a recombinant protein covering the disintegrin-cysteine domain of ADAM9, demonstrated that this receptor directly binds to platelet α6ß1. In vivo studies showed that the interplay between platelet α6ß1 and tumor cell-expressed ADAM9 promotes efficient lung metastasis. The integrin α6ß1-dependent platelet-tumor cell interaction induces platelet activation and favors the extravasation process of tumor cells. Finally, we demonstrate that a pharmacological approach targeting α6ß1 efficiently impairs tumor metastasis through a platelet-dependent mechanism. Our study reveals a mechanism by which platelets promote tumor metastasis and suggests that integrin α6ß1 represents a promising target for antimetastatic therapies.


Asunto(s)
Proteínas ADAM/metabolismo , Integrina alfa6beta1/metabolismo , Proteínas de la Membrana/metabolismo , Metástasis de la Neoplasia , Activación Plaquetaria , Animales , Plaquetas , Comunicación Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Adhesividad Plaquetaria
8.
Thromb Haemost ; 114(6): 1175-88, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26245230

RESUMEN

The ability of cellular fibronectin, found in the vessel wall in a fibrillar conformation, to regulate platelet functions and trigger thrombus formation remains largely unknown. In this study, we evaluated how parietal cellular fibronectin can modulate platelet responses under flow conditions. A fibrillar network was formed by mechanically stretching immobilised dimeric cellular fibronectin. Perfusion of anticoagulated whole blood over this surface resulted in efficient platelet adhesion and thrombus growth. The initial steps of platelet adhesion and activation, as evidenced by filopodia extension and an increase in intracellular calcium levels (419 ± 29 nmol/l), were dependent on integrins α5ß1 and αIIbß3. Subsequent thrombus growth was mediated by these integrins together with the GPIb-V-IX complex, GPVI and Toll-like receptor 4. The involvement of Toll-like receptor 4 could be conveyed via its binding to the EDA region of cellular fibronectin. Upon thrombus formation, the platelets became procoagulant and generated fibrin as revealed by video-microscopy. This work provides evidence that fibrillar cellular fibronectin is a strong thrombogenic surface which supports efficient platelet adhesion, activation, aggregation and procoagulant activity through the interplay of a series of receptors including integrins α5ß1 and αIIbß3, the GPIb-V-IX complex, GPVI and Toll-like receptor 4.


Asunto(s)
Coagulación Sanguínea/fisiología , Fibronectinas/fisiología , Agregación Plaquetaria/fisiología , Animales , Anexina A5/metabolismo , Matriz Extracelular , Fibrina/biosíntesis , Fibroblastos , Fibronectinas/química , Proteínas Inmovilizadas , Integrina beta1/genética , Integrinas/fisiología , Dispositivos Laboratorio en un Chip , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas Analíticas Microfluídicas , Microscopía Electrónica de Rastreo , Adhesividad Plaquetaria , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/deficiencia , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/fisiología , Reología , Estrés Mecánico , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/fisiología
9.
Circulation ; 128(5): 541-52, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23797810

RESUMEN

BACKGROUND: Laminins are major components of basement membranes, well located to interact with platelets upon vascular injury. Laminin-111 (α1ß1γ1) is known to support platelet adhesion but is absent from most blood vessels, which contain isoforms with the α2, α4, or α5 chain. Whether vascular laminins support platelet adhesion and activation and the significance of these interactions in hemostasis and thrombosis remain unknown. METHODS AND RESULTS: Using an in vitro flow assay, we show that laminin-411 (α4ß1γ1), laminin-511 (α5ß1γ1), and laminin-521 (α5ß2γ1), but not laminin-211 (α2ß1γ1), allow efficient platelet adhesion and activation across a wide range of arterial wall shear rates. Adhesion was critically dependent on integrin α6ß1 and the glycoprotein Ib-IX complex, which binds to plasmatic von Willebrand factor adsorbed on laminins. Glycoprotein VI did not participate in the adhesive process but mediated platelet activation induced by α5-containing laminins. To address the significance of platelet/laminin interactions in vivo, we developed a platelet-specific knockout of integrin α6. Platelets from these mice failed to adhere to laminin-411, laminin-511, and laminin-521 but responded normally to a series of agonists. α6ß1-Deficient mice presented a marked decrease in arterial thrombosis in 3 models of injury of the carotid, aorta, and mesenteric arterioles. The tail bleeding time and blood loss remained unaltered, indicating normal hemostasis. CONCLUSIONS: This study reveals an unsuspected important contribution of laminins to thrombus formation in vivo and suggests that targeting their main receptor, integrin α6ß1, could represent an alternative antithrombotic strategy with a potentially low bleeding risk.


Asunto(s)
Adhesión Celular/fisiología , Integrina alfa6beta1/metabolismo , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Trombosis/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Humanos , Integrina alfa6beta1/fisiología , Laminina/fisiología , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Riesgo , Trombosis/patología
10.
Arterioscler Thromb Vasc Biol ; 33(6): 1221-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23559635

RESUMEN

OBJECTIVE: The glycoprotein (GP) Ib-V-IX complex regulates the adhesion, activation, and procoagulant activity of platelets. We previously reported that RAM.1, a rat monoclonal antibody directed against the extracellular domain of mouse GPIbß, diminished adhesion of platelets and chinese hamster ovary cells transfected with the human GPIb-IX complex to von Willebrand factor under flow conditions. Here, we further evaluated the functional importance of GPIbß by studying the impact of RAM.1 on GPIb-mediated platelet responses and in vitro and in vivo thrombus formation. APPROACH AND RESULTS: We show that RAM.1 dramatically reduced GPIb-mediated filopodia extension of chinese hamster ovary GPIb-IX cells after adhesion to von Willebrand factor. RAM.1 also reduced filopodia extension and GPIb-mediated Ca(2+) signaling after adhesion of mouse platelets to von Willebrand factor. RAM.1 inhibited thrombin generation in platelet-rich plasma without impairing phosphatidylserine exposure. In addition, RAM.1 reduced thrombus formation after perfusion of mouse whole blood over collagen in a shear-dependent manner. This effect was confirmed in vivo, because injection of F(ab)'2 fragments of RAM.1 diminished thrombus formation induced by laser beam injury of mesenteric arterioles and forceps injury of the abdominal aorta. In contrast, RAM.1 F(ab)'2 did not prolong the tail-bleeding time or increase the volume of blood lost. CONCLUSIONS: These findings are the first evidence that targeting a subunit other than GPIbα can lead to an antithrombotic effect via the GPIb-V-IX complex. This could represent an alternative way to reduce thrombus formation with a minor impact on hemostasis.


Asunto(s)
Arteriopatías Oclusivas/prevención & control , Adhesividad Plaquetaria/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Trombosis/prevención & control , Factor de von Willebrand/metabolismo , Animales , Arteriopatías Oclusivas/fisiopatología , Tiempo de Sangría , Adhesión Celular/fisiología , Cricetinae , Modelos Animales de Enfermedad , Humanos , Ratones , Adhesividad Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Distribución Aleatoria , Ratas , Sensibilidad y Especificidad , Transducción de Señal , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Trombosis/fisiopatología
11.
J Pharmacol Exp Ther ; 341(1): 156-63, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238212

RESUMEN

Glycoprotein VI (GPVI) has been proposed as a promising antiplatelet target, because its blockade prevents experimental thrombosis without impairing hemostasis. The objective of this study was to develop a preclinical tool to evaluate the role of human GPVI (hGPVI) in various models of thrombosis and to screen anti-GPVI compounds. A genetically modified mouse strain expressing hGPVI has been developed using a knockin strategy. The mice were viable and fertile and did not present any hematological defects. Approximately 3700 copies of human GPVI were detected at the platelet surface. Platelet aggregation, fibrinogen binding, and P-selectin exposure were normal in response to various agonists. The 9O12.2 Fab fragment directed against human GPVI bound to hGPVI platelets in vitro and ex vivo and markedly reduced collagen- and collagen-related peptide-induced responses. Injection of 9O12.2 into hGPVI animals did not prolong the tail bleeding time but provided protection against lethal thromboembolism induced by a collagen/adrenaline mixture. In addition, 9O12.2 reduced arterial thrombus growth by 44% after superficial laser injury, 43% after deep laser injury in mice pretreated with hirudin, and 48% after mechanical injury. In conclusion, we have developed a humanized mouse model that could be used in preclinical studies to evaluate the effects of anti-GPVI compounds.


Asunto(s)
Modelos Animales de Enfermedad , Fibrinolíticos/uso terapéutico , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/genética , Trombosis/genética , Trombosis/metabolismo , Animales , Fibrinolíticos/administración & dosificación , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Transgénicos , Glicoproteínas de Membrana Plaquetaria/fisiología , Trombosis/tratamiento farmacológico , Resultado del Tratamiento
12.
Arterioscler Thromb Vasc Biol ; 31(1): 117-24, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20651280

RESUMEN

OBJECTIVE: The identification of platelet-reactive proteins exclusively present in atherosclerotic plaques could provide interesting targets for effective and safe antithrombotic strategies. In this context, we explored platelet adhesion and activation to tenascin-C (TN-C), a matrix protein preferentially found within atheroma. METHODS AND RESULTS: We show that platelets efficiently adhere to TN-C under both static and flow conditions. Videomicroscopy revealed a unique behavior under flow, with platelets exhibiting stationary adhesion to TN-C; in contrast, platelets rolled over von Willebrand factor and detached from fibrinogen. Platelet interaction with TN-C was predominantly supported by integrin α(2)ß(1) under static conditions, whereas under high shear, it was dependent on both the α(2)ß(1) integrin and the glycoprotein Ib-IX complex. Integrin α(IIb)ß(3) appeared to play a secondary role but only at low shear rates. The glycoprotein Ib-IX-dependent interaction was indirect, relying on von Willebrand factor, and increased as a function of wall shear rate. Von Willebrand factor bound directly to TN-C, as shown by ELISA and coimmunoprecipitation, suggesting that it acts as a bridge between TN-C and platelets. The adhesion of platelets to TN-C triggered their activation, as demonstrated by a shape change and increases in intracellular calcium level. CONCLUSIONS: This study provides evidence that TN-C serves as a novel adhesive matrix for platelets in a context that is relevant to atherothrombosis.


Asunto(s)
Aterosclerosis/sangre , Plaquetas/metabolismo , Activación Plaquetaria , Tenascina/metabolismo , Aterosclerosis/fisiopatología , Calcio/sangre , Forma de la Célula , Fibronectinas/metabolismo , Humanos , Integrina alfa2/sangre , Integrina alfa2beta1/sangre , Integrina beta3/sangre , Glicoproteínas de Membrana/sangre , Microscopía por Video , Adhesividad Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Tiempo , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...