Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38060311

RESUMEN

Platelets from patients with myeloproliferative neoplasms (MPNs) exhibit a hyperreactive phenotype. Here, we found elevated P-selectin exposure and platelet-leukocyte aggregates indicating activation of platelets from essential thrombocythemia (ET) patients. Single-cell RNA-seq analysis of primary samples revealed significant enrichment of transcripts related to platelet activation, mTOR, and oxidative phosphorylation in ET patient platelets. These observations were validated via proteomic profiling. Platelet metabolomics revealed distinct metabolic phenotypes consisting of elevated ATP generation accompanied by increases in the levels of multiple intermediates of the tricarboxylic acid cycle, but lower α-ketoglutarate (α-KG) in MPN patients. Inhibition of PI3K/AKT/mTOR signaling significantly reduced metabolic responses and hyperreactivity in MPN patient platelets, while α-KG supplementation markedly reduced oxygen consumption and ATP generation. Ex vivo incubation of platelets from both MPN patients and Jak2 V617F-knockin mice with α-KG supplementation significantly reduced platelet activation responses. Oral α-KG supplementation of Jak2 V617F mice decreased splenomegaly and reduced hematocrit, monocyte, and platelet counts. Finally, α-KG treatment significantly decreased proinflammatory cytokine secretion from MPN CD14+ monocytes. Our results reveal a previously unrecognized metabolic disorder in conjunction with aberrant PI3K/AKT/mTOR signaling that contributes to platelet hyperreactivity in MPN patients.


Asunto(s)
Trastornos Mieloproliferativos , Neoplasias , Trombocitemia Esencial , Humanos , Ratones , Animales , Multiómica , Fosfatidilinositol 3-Quinasas/genética , Proteómica , Proteínas Proto-Oncogénicas c-akt/genética , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trombocitemia Esencial/genética , Inflamación , Serina-Treonina Quinasas TOR/genética , Adenosina Trifosfato , Janus Quinasa 2/genética , Mutación
2.
Arterioscler Thromb Vasc Biol ; 44(2): e39-e53, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38126172

RESUMEN

BACKGROUND: Platelet-rich thrombi occlude arteries causing fatal infarcts like heart attacks and strokes. Prevention of thrombi by current antiplatelet agents can cause major bleeding. Instead, we propose using N-acetyl cysteine (NAC) to act against the protein VWF (von Willebrand factor), and not platelets, to prevent arterial thrombi from forming. METHODS: NAC was assessed for its ability to prevent arterial thrombosis by measuring platelet accumulation rate and occlusion time using a microfluidic model of arterial thrombosis with human blood. Acute clot formation, clot stability, and tail bleeding were measured in vivo with the murine modified Folts model. The effect of NAC in the murine model after 6 hours was also measured to determine any persistent effects of NAC after it has been cleared from the blood. RESULTS: We demonstrate reduction of thrombi formation following treatment with NAC in vitro and in vivo. Human whole blood treated with 3 or 5 mmol/L NAC showed delayed thrombus formation 2.0× and 3.7× longer than control, respectively (P<0.001). Blood treated with 10 mmol/L NAC did not form an occlusive clot, and no macroscopic platelet aggregation was visible (P<0.001). In vivo, a 400-mg/kg dose of NAC prevented occlusive clots from forming in mice without significantly affecting tail bleeding times. A lower dose of NAC significantly reduced clot stability. Mice given multiple injections showed that NAC has a lasting and cumulative effect on clot stability, even after being cleared from the blood (P<0.001). CONCLUSIONS: Both preclinical models demonstrate that NAC prevents thrombus formation in a dose-dependent manner without significantly affecting bleeding time. This work highlights a new pathway for preventing arterial thrombosis, different from antiplatelet agents, using an amino acid derivative as an antithrombotic therapeutic.


Asunto(s)
Tromboembolia , Trombosis , Ratones , Humanos , Animales , Inhibidores de Agregación Plaquetaria/farmacología , Acetilcisteína/farmacología , Trombosis/inducido químicamente , Trombosis/prevención & control , Trombosis/tratamiento farmacológico , Agregación Plaquetaria , Plaquetas/metabolismo , Hemorragia/metabolismo , Factor de von Willebrand/metabolismo
3.
Blood Adv ; 7(13): 3180-3191, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-36884296

RESUMEN

This report identifies a novel variant form of the inherited bleeding disorder Glanzmann thrombasthenia, exhibiting only mild bleeding in a physically active individual. The platelets cannot aggregate ex vivo with physiologic agonists of activation, although microfluidic analysis with whole blood displays moderate ex vivo platelet adhesion and aggregation consistent with mild bleeding. Immunocytometry shows reduced expression of αIIbß3 on quiescent platelets that spontaneously bind/store fibrinogen, and activation-dependent antibodies (ligand-induced binding site-319.4 and PAC-1) report ß3 extension suggesting an intrinsic activation phenotype. Genetic analysis reveals a single F153Sß3 substitution within the ßI-domain from a heterozygous T556C nucleotide substitution of ITGB3 exon 4 in conjunction with a previously reported IVS5(+1)G>A splice site mutation with undetectable platelet messenger RNA accounting for hemizygous expression of S153ß3. F153 is completely conserved among ß3 of several species and all human ß-integrin subunits suggesting that it may play a vital role in integrin structure/function. Mutagenesis of αIIb-F153Sß3 also displays reduced levels of a constitutively activated αIIb-S153ß3 on HEK293T cells. The overall structural analysis suggests that a bulky aromatic, nonpolar amino acid (F,W)153ß3 is critical for maintaining the resting conformation of α2- and α1-helices of the ßI-domain because small amino acid substitutions (S,A) facilitate an unhindered inward movement of the α2- and α1-helices of the ßI-domain toward the constitutively active αIIbß3 conformation, while a bulky aromatic, polar amino acid (Y) hinders such movements and restrains αIIbß3 activation. The data collectively demonstrate that disruption of F153ß3 can significantly alter normal integrin/platelet function, although reduced expression of αIIb-S153ß3 may be compensated by a hyperactive conformation that promotes viable hemostasis.


Asunto(s)
Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Trombastenia , Humanos , Aminoácidos/genética , Células HEK293 , Mutación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombastenia/genética , Trombastenia/metabolismo
5.
Nat Commun ; 14(1): 850, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792666

RESUMEN

Sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1 (SVEP1) is an extracellular matrix protein that causally promotes vascular disease and associates with platelet reactivity in humans. Here, using a human genomic and proteomic approach, we identify a high affinity, disease-relevant, and potentially targetable interaction between SVEP1 and the orphan receptor Platelet and Endothelial Aggregation Receptor 1 (PEAR1). This interaction promotes PEAR1 phosphorylation and disease associated AKT/mTOR signaling in vascular cells and platelets. Mice lacking SVEP1 have reduced platelet activation, and exogenous SVEP1 induces PEAR1-dependent activation of platelets. SVEP1 and PEAR1 causally and concordantly relate to platelet phenotypes and cardiovascular disease in humans, as determined by Mendelian Randomization. Targeting this receptor-ligand interaction may be a viable therapeutic strategy to treat or prevent cardiovascular and thrombotic disease.


Asunto(s)
Plaquetas , Proteómica , Humanos , Animales , Ratones , Plaquetas/metabolismo , Ligandos , Receptores de Superficie Celular/metabolismo , Agregación Plaquetaria , Moléculas de Adhesión Celular/metabolismo
6.
Blood ; 139(14): 2240-2251, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35143643

RESUMEN

von Willebrand factor (VWF) plays a key role in normal hemostasis, and deficiencies of VWF lead to clinically significant bleeding. We sought to identify novel modifiers of VWF levels in endothelial colony-forming cells (ECFCs) using single-cell RNA sequencing (scRNA-seq). ECFCs were isolated from patients with low VWF levels (plasma VWF antigen levels between 30 and 50 IU/dL) and from healthy controls. Human umbilical vein endothelial cells were used as an additional control cell line. Cells were characterized for their Weibel Palade body (WPB) content and VWF release. scRNA-seq of all cell lines was performed to evaluate for gene expression heterogeneity and for candidate modifiers of VWF regulation. Candidate modifiers identified by scRNA-seq were further characterized with small-interfering RNA (siRNA) experiments to evaluate for effects on VWF. We observed that ECFCs derived from patients with low VWF demonstrated alterations in baseline WPB metrics and exhibit impaired VWF release. scRNA-seq analyses of these endothelial cells revealed overall decreased VWF transcription, mosaicism of VWF expression, and genes that are differentially expressed in low VWF ECFCs and control endothelial cells (control ECs). An siRNA screen of potential VWF modifiers provided further evidence of regulatory candidates, and 1 such candidate, FLI1, alters the transcriptional activity of VWF. In conclusion, ECFCs from individuals with low VWF demonstrate alterations in their baseline VWF packaging and release compared with control ECs. scRNA-seq revealed alterations in VWF transcription, and siRNA screening identified multiple candidate regulators of VWF.


Asunto(s)
Enfermedades de von Willebrand , Factor de von Willebrand , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Análisis de la Célula Individual , Cuerpos de Weibel-Palade/metabolismo , Enfermedades de von Willebrand/metabolismo , Factor de von Willebrand/metabolismo
7.
Nanomedicine ; 35: 102405, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33932591

RESUMEN

Platelet accumulation by VWF under high shear rates at the site of atherosclerotic plaque rupture leads to myocardial infarction and stroke. Current anti-platelet therapies remain ineffective for a large percentage of the population, while presenting significant risks for bleeding. We explore a novel way to inhibit arterial thrombus formation. Theoretically, a negative charge may influence the tertiary structure of VWF to favor the globular configuration by biophysical means without the use of platelet inactivating drugs. We tested this hypothesis experimentally for charged nanoparticles (CNPs) to inhibit thrombus formation in a microfluidic thrombosis assay (MTA). Several different CNPs demonstrated the ability to retard thrombotic occlusion in the MTA. A preliminary study in mice shows that thrombus stability is weaker with CNP administration and bleeding times are not markedly prolonged. The CNPs tested here show promise as a new class of antithrombotic therapies that act by biophysical means rather than biochemical pathways.


Asunto(s)
Plaquetas/metabolismo , Fibrinolíticos , Técnicas Analíticas Microfluídicas , Nanopartículas , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Trombosis , Animales , Tiempo de Sangría , Fibrinolíticos/química , Fibrinolíticos/uso terapéutico , Humanos , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Trombosis/tratamiento farmacológico , Trombosis/metabolismo
8.
Cell Mol Bioeng ; 13(4): 379-390, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32952737

RESUMEN

INTRODUCTION: Pathological flows in patients with severe aortic stenosis are associated with acquired von Willebrand syndrome. This syndrome is characterized by excessive cleavage of von Willebrand factor by its main protease, A Disintegrin and Metalloproteinase with a Thrombospondin Type 1 Motif, Member 13 (ADAMTS13) leading to decreased VWF function and mucocutaneous bleeding. Aortic valve replacement and correction of the flow behavior to physiological levels reverses the syndrome, supporting the association between pathological flow and acquired von Willebrand syndrome. We investigated the effects of shear and elongational rates on von Willebrand factor cleavage in the presence of ADAMTS13. METHODS: We identified acquired von Willebrand syndrome in five patients with severe aortic stenosis. Doppler echography values from these patients were used to develop three computational fluid dynamic (CFD) aortic valve models (normal, mild and severe stenosis). Shear, elongational rates and exposure times identified in the CFD simulations were used as parameters for the design of microfluidic devices to test the effects of pathologic shear and elongational rates on the structure and function of von Willebrand factor. RESULTS: The shear rates (0-10,000s-1), elongational rates (0-1000 s-1) and exposure times (1-180 ms) tested in our microfluidic designs mimicked the flow features identified in patients with aortic stenosis. The shear and elongational rates tested in vitro did not lead to excessive cleavage or decreased function of von Willebrand factor in the presence of the protease. CONCLUSIONS: High shear and elongational rates in the presence of ADAMTS13 are not sufficient for excessive cleavage of von Willebrand Factor.

9.
Blood Adv ; 4(14): 3258-3267, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32697818

RESUMEN

von Willebrand factor (VWF) is essential for the induction of arterial thrombosis. In this study, we investigated the critical role of platelet VWF in occlusive thrombosis formation at high shear in mice that do not express platelet VWF (Nbeal2-/-). Using in silico modeling, in vitro high-shear microfluidics, and an in vivo Folts model of arterial thrombosis we reproduced the platelet dynamics that occur under pathological flow in a stenosed vessel. Computational fluid dynamics (CFDs) simulated local hemodynamics in a stenosis based on arterial geometries. The model predicted shear rates, time course of platelet adhesion, and time to occlusion. These predictions were validated in vitro and in vivo. Occlusive thrombosis developed in wild-type control mice that had normal levels of plasma VWF and platelet VWF in vitro and in vivo. Occlusive thrombosis did not form in the Nbeal2-/- mice that had normal plasma VWF and an absence of platelet VWF. Occlusive thrombosis was corrected in Nbeal2-/- microfluidic assays by the addition of exogenous normal platelets with VWF. Combining model and experimental data, we demonstrated the necessary requirement of platelet VWF in α-granules in forming an occlusive thrombus under high shear. These results could inspire new pharmacological targets specific to pathological conditions and prevent arterial thrombosis.


Asunto(s)
Trombosis , Enfermedades Vasculares , Animales , Plaquetas , Proteínas Sanguíneas , Ratones , Adhesividad Plaquetaria , Factor de von Willebrand/genética
10.
Arterioscler Thromb Vasc Biol ; 39(9): 1831-1842, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31291760

RESUMEN

Objective- Acquired von Willebrand syndrome is defined by excessive cleavage of the VWF (von Willebrand Factor) and is associated with impaired primary hemostasis and severe bleeding. It often develops when blood is exposed to nonphysiological flow such as in aortic stenosis or mechanical circulatory support. We evaluated the role of laminar, transitional, and turbulent flow on VWF cleavage and the effects on VWF function. Approach and Results- We used a vane rheometer to generate laminar, transitional, and turbulent flow and evaluate the effect of each on VWF cleavage in the presence of ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type-1 motif, member 13). We performed functional assays to evaluate the effect of these flows on VWF structure and function. Computational fluid dynamics was used to estimate the flow fields and forces within the vane rheometer under each flow condition. Turbulent flow is required for excessive cleavage of VWF in an ADAMTS13-dependent manner. The assay was repeated with whole blood, and the turbulent flow had the same effect. Our computational fluid dynamics results show that under turbulent conditions, the Kolmogorov scale approaches the size of VWF. Finally, cleavage of VWF in this study has functional consequences under flow as the resulting VWF has decreased ability to bind platelets and collagen. Conclusions- Turbulent flow mediates VWF cleavage in the presence of ADAMTS13, decreasing the ability of VWF to sustain platelet adhesion. These findings impact the design of mechanical circulatory support devices and are relevant to pathological environments where turbulence is added to circulation.


Asunto(s)
Proteína ADAMTS13/fisiología , Factor de von Willebrand/química , Adulto , Colágeno/química , Humanos , Hidrodinámica , Masculino , Persona de Mediana Edad , Adhesividad Plaquetaria , Resistencia al Corte , Factor de von Willebrand/fisiología
11.
Blood ; 134(9): 727-740, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31311815

RESUMEN

Aging and chronic inflammation are independent risk factors for the development of atherothrombosis and cardiovascular disease. We hypothesized that aging-associated inflammation promotes the development of platelet hyperreactivity and increases thrombotic risk during aging. Functional platelet studies in aged-frail adults and old mice demonstrated that their platelets are hyperreactive and form larger thrombi. We identified tumor necrosis factor α (TNF-α) as the key aging-associated proinflammatory cytokine responsible for platelet hyperreactivity. We further showed that platelet hyperreactivity is neutralized by abrogating signaling through TNF-α receptors in vivo in a mouse model of aging. Analysis of the bone marrow compartments showed significant platelet-biased hematopoiesis in old mice reflected by increased megakaryocyte-committed progenitor cells, megakaryocyte ploidy status, and thrombocytosis. Single-cell RNA-sequencing analysis of native mouse megakaryocytes showed significant reprogramming of inflammatory, metabolic, and mitochondrial gene pathways in old mice that appeared to play a significant role in determining platelet hyperreactivity. Platelets from old mice (where TNF-α was endogenously increased) and from young mice exposed to exogenous TNF-α exhibited significant mitochondrial changes characterized by elevated mitochondrial mass and increased oxygen consumption during activation. These mitochondrial changes were mitigated upon TNF-α blockade. Similar increases in platelet mitochondrial mass were seen in platelets from patients with myeloproliferative neoplasms, where TNF-α levels are also increased. Furthermore, metabolomics studies of platelets from young and old mice demonstrated age-dependent metabolic profiles that may differentially poise platelets for activation. Altogether, we present previously unrecognized evidence that TNF-α critically regulates megakaryocytes resident in the bone marrow niche and aging-associated platelet hyperreactivity and thrombosis.


Asunto(s)
Envejecimiento , Plaquetas/inmunología , Inflamación/inmunología , Mitocondrias/inmunología , Trombosis/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Plaquetas/patología , Inflamación/patología , Megacariocitos/inmunología , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/patología , Activación Plaquetaria , Trombosis/patología
12.
Res Pract Thromb Haemost ; 2(2): 380-389, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30046742

RESUMEN

BACKGROUND: Antiphospholipid syndrome (APS) is characterized by recurrent thromboembolic events in the setting of pathologic autoantibodies, some of which are directed to ß2-Glycoprotein 1 (ß2GPI). The mechanisms of thrombosis in APS appear to be multifactorial and likely include a component of endothelial activation. Among other things, activated endothelium secretes von Willebrand factor, a hemostatic protein that in excess can increase the risk of thrombosis. OBJECTIVE: We hypothesized that anti-ß2GPI antibodies could regulate the release and modulation of VWF from endothelial cells. PATIENTS/METHODS: Isolated anti-ß2GPI antibodies from patients with APS were assayed for their ability to induced VWF release from HUVECs and modulate the effects of ADAMTS13 in a shear-dependent assay. RESULTS: We observed that anti-ß2GPI antibodies from some patients with APS induced VWF release from human endothelial cells but did not induce formation of cell-anchored VWF-platelet strings. Finally, we also determined that one of the Anti-ß2GPI antibodies tested can inhibit the function of ADAMTS13, the main modulator of extracellular VWF. CONCLUSIONS: These results suggest that VWF and ADAMTS13 may play a role in the prothrombotic phenotype of APS.

13.
Sci Transl Med ; 9(409)2017 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-28954929

RESUMEN

Gut ischemia is common in critically ill patients, promoting thrombosis and inflammation in distant organs. The mechanisms linking hemodynamic changes in the gut to remote organ thrombosis remain ill-defined. We demonstrate that gut ischemia in the mouse induces a distinct pulmonary thrombotic disorder triggered by neutrophil macroaggregates. These neutrophil aggregates lead to widespread occlusion of pulmonary arteries, veins, and the microvasculature. A similar pulmonary neutrophil-rich thrombotic response occurred in humans with the acute respiratory distress syndrome. Intravital microscopy during gut ischemia-reperfusion injury revealed that rolling neutrophils extract large membrane fragments from remnant dying platelets in multiple organs. These platelet fragments bridge adjacent neutrophils to facilitate macroaggregation. Platelet-specific deletion of cyclophilin D, a mitochondrial regulator of cell necrosis, prevented neutrophil macroaggregation and pulmonary thrombosis. Our studies demonstrate the existence of a distinct pulmonary thrombotic disorder triggered by dying platelets and neutrophil macroaggregates. Therapeutic targeting of platelet death pathways may reduce pulmonary thrombosis in critically ill patients.


Asunto(s)
Tracto Gastrointestinal/irrigación sanguínea , Tracto Gastrointestinal/patología , Isquemia/complicaciones , Pulmón/patología , Neutrófilos/patología , Trombosis/etiología , Trombosis/patología , Animales , Plaquetas/metabolismo , Agregación Celular , Membrana Celular/metabolismo , Peptidil-Prolil Isomerasa F , Ciclofilinas/deficiencia , Ciclofilinas/metabolismo , Tracto Gastrointestinal/fisiopatología , Humanos , Isquemia/fisiopatología , Pulmón/irrigación sanguínea , Pulmón/fisiopatología , Ratones Endogámicos C57BL , Fosfatidilserinas/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/fisiopatología , Circulación Esplácnica
14.
Blood ; 121(22): 4555-66, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23550035

RESUMEN

Thrombosis promotes leukocyte infiltration into inflamed tissues, leading to organ injury in a broad range of diseases; however, the mechanisms by which thrombi guide leukocytes to sites of vascular injury remain ill-defined. Using mouse models of endothelial injury (traumatic or ischemia reperfusion), we demonstrate a distinct process of leukocyte recruitment, termed "directed intravascular migration," specifically mediated by platelet thrombi. Single adherent platelets and platelet aggregates stimulated leukocyte shape change at sites of endothelial injury; however, only thrombi were capable of inducing directed intravascular leukocyte migration. Leukocyte recruitment and migration induced by platelet thrombi occurred most prominently in veins but could also occur in arteries following ischemia-reperfusion injury. In vitro studies demonstrated a major role for platelet-derived NAP-2 (CXCL-7) and its CXCR1/2 receptor in regulating leukocyte polarization and motility. In vivo studies demonstrated the presence of an NAP-2 chemotactic gradient within the thrombus body. Pharmacologic blockade of CXCR1/2 as well as genetic deletion of NAP-2 markedly reduced leukocyte shape change and intrathrombus migration. These studies define a distinct process of leukocyte migration that is initiated by homotypic adhesive interactions between platelets, leading to the development of an NAP-2 chemotactic gradient within the thrombus body that guides leukocytes to sites of vascular injury.


Asunto(s)
Plaquetas/citología , Quimiocinas CXC/metabolismo , Leucocitos/citología , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Trombosis/inmunología , Animales , Plaquetas/inmunología , Plaquetas/metabolismo , Adhesión Celular/inmunología , Movimiento Celular/inmunología , Polaridad Celular/inmunología , Proteínas Fluorescentes Verdes/genética , Leucocitos/inmunología , Arterias Mesentéricas/inmunología , Arterias Mesentéricas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Lesiones por Pinchazo de Aguja/inmunología , Lesiones por Pinchazo de Aguja/patología , Neutrófilos/citología , Neutrófilos/inmunología , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología
15.
Blood ; 117(9): 2718-27, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21156842

RESUMEN

Platelets have evolved a highly specialized membrane skeleton that provides stability to the plasma membrane and facilitates adhesion under high shear stress. The cytoskeletal anchorage of glycoprotein (GP) Ibα plays an important role in regulating the membrane skeleton. However, its role in regulating membrane stability remains unknown. To investigate this role, we have developed a new mouse model that expresses wild-type human GPIbα (hGPIbα(WT)), or a mutant form of human GPIbα that has a selective defect in its ability to bind filamin A and anchor to the membrane skeleton (hGPIbα(FW)-Phe568Ala and Trp570Ala substitutions). Our study demonstrates that the link between platelet GPIb and the cytoskeleton does not alter the intrinsic ligand binding function of GPIbα or the ability of the receptor to stimulate integrin α(IIb)ß(3)-dependent spreading. However, exposure of hGPIbα(FW) platelets to pathologic shear rate levels (5000 to 40,000 s(-1)) leads to the development of unstable membrane tethers, defective platelet adhesion, and loss of membrane integrity, leading to complete disintegration of the platelet cell body. These outcomes suggest that the GPIbα-filamin A interaction not only regulates the architecture of the membrane skeleton, but also maintains the mechanical stability of the plasma membrane under conditions of high shear.


Asunto(s)
Plaquetas/citología , Membrana Celular/metabolismo , Proteínas Contráctiles/metabolismo , Proteínas de Microfilamentos/metabolismo , Adhesividad Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Estrés Mecánico , Animales , Plaquetas/efectos de los fármacos , Caspasas/metabolismo , Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Filaminas , Humanos , Proteínas Inmovilizadas/farmacología , Ratones , Ratones Transgénicos , Modelos Animales , Proteínas Mutantes/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Factor de von Willebrand/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...