Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Blood ; 132(24): e35-e46, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30275110

RESUMEN

Antithrombotic therapies reduce cardiovascular diseases by preventing arterial thrombosis and thromboembolism, but at expense of increased bleeding risks. Arterial thrombosis studies using genetically modified mice have been invaluable for identification of new molecular targets. Because of low sample sizes and heterogeneity in approaches or methodologies, a formal meta-analysis to compare studies of mice with single-gene defects encountered major limitations. To overcome these, we developed a novel synthesis approach to quantitatively scale 1514 published studies of arterial thrombus formation (in vivo and in vitro), thromboembolism, and tail-bleeding of genetically modified mice. Using a newly defined consistency parameter (CP), indicating the strength of published data, comparisons were made of 431 mouse genes, of which 17 consistently contributed to thrombus formation without affecting hemostasis. Ranking analysis indicated high correlations between collagen-dependent thrombosis models in vivo (FeCl3 injury or ligation/compression) and in vitro. Integration of scores and CP values resulted in a network of protein interactions in thrombosis and hemostasis (PITH), which was combined with databases of genetically linked human bleeding and thrombotic disorders. The network contained 2946 nodes linked to modifying genes of thrombus formation, mostly with expression in megakaryocytes. Reactome pathway analysis and network characteristics revealed multiple novel genes with potential contribution to thrombosis/hemostasis. Studies with additional knockout mice revealed that 4 of 8 (Apoe, Fpr2, Ifnar1, Vps13a) new genes were modifying in thrombus formation. The PITH network further: (i) revealed a high similarity of murine and human hemostatic and thrombotic processes and (ii) identified multiple new candidate proteins regulating these processes.


Asunto(s)
Hemorragia , Trombosis , Animales , Modelos Animales de Enfermedad , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Ratones , Ratones Noqueados , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología
2.
Platelets ; 25(1): 71-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23301869

RESUMEN

Early ischemic ventricular fibrillation (VF) in the setting of an acute myocardial infarction (AMI) due to thrombotic coronary occlusion remains a major health problem. Several animal studies have shown that platelet-dense granule contents released during thrombus formation can induce arrhythmias. We hypothesize that the platelet release reaction is involved in the predisposition to early ischemic VF. A case-control study was performed in patients who survived VF during a first AMI ("cases," n = 26) and in patients with one previous AMI without arrhythmias ("controls," n = 24). All patients were on aspirin 100 mg OD. Baseline platelet activation was assessed with flow cytometry. Response to activation was assessed with aggregometry, flow cytometry and PFA-100 analysis. Differences in platelet contents and content release were assessed by labeling platelet-dense granules with mepacrine and by measuring serotonin and ADP/ATP content. Patient and infarct characteristics and baseline platelet function tests were similar between groups. The mean time from event was 4.9 (±3.2) years among cases and 4.7 (±2.7) years among controls. Dense granule release was similar in cases versus controls. Platelet serotonin content in cases was higher than in controls (611 ± 118 ng/10E(9) platelets vs. 536 ± 141 ng/10(9), p = 0.048). Even years after the event, elevations in the platelet dense granule contents between VF survivors and controls may be detected. These preliminary findings shed new light on the pathophysiological mechanisms underlying ischemic VF, as platelet-dense granules may contain mediators of early ischemic VF risk.


Asunto(s)
Plaquetas/patología , Fibrilación Ventricular/sangre , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sobrevivientes , Fibrilación Ventricular/patología
3.
Haematologica ; 98(11): 1810-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23753027

RESUMEN

Initial platelet arrest at the exposed arterial vessel wall is mediated through glycoprotein Ibα binding to the A1 domain of von Willebrand factor. This interaction occurs at sites of elevated shear force, and strengthens upon increasing hydrodynamic drag. The increased interaction requires shear-dependent exposure of the von Willebrand factor A1 domain, but the contribution of glycoprotein Ibα remains ill defined. We have previously found that glycoprotein Ibα forms clusters upon platelet cooling and hypothesized that such a property enhances the interaction with von Willebrand factor under physiological conditions. We analyzed the distribution of glycoprotein Ibα with Förster resonance energy transfer using time-gated fluorescence lifetime imaging microscopy. Perfusion at a shear rate of 1,600 s(-1) induced glycoprotein Ibα clusters on platelets adhered to von Willebrand factor, while clustering did not require von Willebrand factor contact at 10,000 s(-1). Shear-induced clustering was reversible, not accompanied by granule release or αIIbß3 activation and improved glycoprotein Ibα-dependent platelet interaction with von Willebrand factor. Clustering required glycoprotein Ibα translocation to lipid rafts and critically depended on arachidonic acid-mediated binding of 14-3-3ζ to its cytoplasmic tail. This newly identified mechanism emphasizes the ability of platelets to respond to mechanical force and provides new insights into how changes in hemodynamics influence arterial thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Resistencia al Corte/fisiología , Factor de von Willebrand/metabolismo , Adhesión Celular/fisiología , Análisis por Conglomerados , Humanos , Unión Proteica/fisiología , Distribución Aleatoria
5.
Haematologica ; 97(8): 1149-57, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22491740

RESUMEN

BACKGROUND: In normal platelets, insulin inhibits agonist-induced Ca(2+) mobilization by raising cyclic AMP. Platelet from patients with type 2 diabetes are resistant to insulin and show increased Ca(2+) mobilization, aggregation and procoagulant activity. We searched for the cause of this insulin resistance. DESIGN AND METHODS: Platelets, the megakaryocytic cell line CHRF-288-11 and primary megakaryocytes were incubated with adipokines and with plasma from individuals with a disturbed adipokine profile. Thrombin-induced Ca(2+) mobilization and signaling through the insulin receptor and insulin receptor substrate 1 were measured. Abnormalities induced by adipokines were compared with abnormalities found in platelets from patients with type 2 diabetes. RESULTS: Resistin, leptin, plasminogen activator inhibitor-1 and retinol binding protein 4 left platelets unchanged but induced insulin resistance in CHRF-288-11 cells. Interleukin-6, tumor necrosis factor-α and visfatin had no effect. These results were confirmed in primary megakaryocytes. Contact with adipokines for 2 hours disturbed insulin receptor substrate 1 Ser(307)-phosphorylation, while contact for 72 hours caused insulin receptor substrate 1 degradation. Plasma with a disturbed adipokine profile also made CHRF-288-11 cells insulin-resistant. Platelets from patients with type 2 diabetes showed decreased insulin receptor substrate 1 expression. CONCLUSIONS: Adipokines resistin, leptin, plasminogen activator-1 and retinol binding protein 4 disturb insulin receptor substrate 1 activity and expression in megakaryocytes. This might be a cause of the insulin resistance observed in platelets from patients with type 2 diabetes.


Asunto(s)
Resistencia a la Insulina , Leptina/metabolismo , Megacariocitos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Resistina/metabolismo , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Adipoquinas/metabolismo , Plaquetas/metabolismo , Calcio/metabolismo , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Síndrome Metabólico/metabolismo
6.
Haematologica ; 97(10): 1514-22, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22371179

RESUMEN

BACKGROUND: Cold storage of platelets reduces bacterial growth and preserves their hemostatic properties better than current procedures do. However, storage at 0°C induces [14-3-3ζ-glycoprotein Ibα] association, 14-3-3ζ release from phospho-Bad, Bad activation and apoptosis. DESIGN AND METHODS: We investigated whether arachidonic acid, which also binds 14-3-3ζ, contributes to coldinduced apoptosis. RESULTS: Cold storage activated P38-mitogen-activated protein kinase and released arachidonic acid, which accumulated due to cold inactivation of cyclooxygenase-1/thromboxane synthase. Accumulated arachidonic acid released 14-3-3ζ from phospho-Bad and decreased the mitochondrial membrane potential, which are steps in the induction of apoptosis. Addition of arachidonic acid did the same and its depletion made platelets resistant to cold-induced apoptosis. Incubation with biotin-arachidonic acid revealed formation of an [arachidonic acid-14-3-3ζ-glycoprotein Ibα] complex. Indomethacin promoted complex formation by accumulating arachidonic acid and released 14-3-3ζ from cyclo-oxygenase-1. Arachidonic acid depletion prevented the cold-induced reduction of platelet survival in mice. CONCLUSIONS: We conclude that cold storage induced apoptosis through an [arachidonic acid-14-3-3ζ-glycoprotein Ibα] complex, which released 14-3-3ζ from Bad in an arachidonic acid-dependent manner. Although arachidonic acid depletion reduced agonist-induced thromboxane A(2) formation and aggregation, arachidonic acid repletion restored these functions, opening ways to reduce apoptosis during storage without compromising hemostatic functions post-transfusion.


Asunto(s)
Proteínas 14-3-3/metabolismo , Ácido Araquidónico/fisiología , Plaquetas , Conservación de la Sangre , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ácido Araquidónico/metabolismo , Ácido Araquidónico/farmacología , Plaquetas/metabolismo , Supervivencia Celular , Frío , Ciclooxigenasa 1/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Activación Plaquetaria , Unión Proteica/efectos de los fármacos , Proteína Letal Asociada a bcl/metabolismo
7.
Platelets ; 23(8): 617-25, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22273509

RESUMEN

The main responses of P2Y(1) ligation are platelet shape change and transient aggregation while P2Y(12) ligation amplifies P2Y(1)-induced aggregation and accelerates aggregation, secretion and thromboxane A(2) production induced by other agonist-receptor complexes. We searched for new targets of P2Y signalling using micro-arrays with 144 peptides representing known phosphosites of protein tyrosine kinases. ADP induced phosphorylation of peptides representing surface receptors, second messenger enzymes and cytoskeletal proteins. Strong phosphorylation was found in peptides representing Ephrin-receptor family members. Blockade of P2Y(1/12) inhibited phosphorylation of EphA4- and EphB1-peptides on micro-arrays. The EphA2/4 inhibitor 2,5-dimethylpyrrolyl benzoic acid derivative interfered with P2Y(1/12)-induced EphA4 phosphorylation, left P2Y(1)-induced aggregation unchanged but inhibited with P2Y(12)-induced secretion, second phase aggregation and thrombus formation on collagen at 1600 s(-1). These results show that platelet EphA4 is an important intermediate in P2Y(12)-induced granule secretion.


Asunto(s)
Plaquetas/enzimología , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptor EphA4/agonistas , Receptores Purinérgicos P2Y12/metabolismo , Vesículas Secretoras/enzimología , Adenosina Difosfato/farmacología , Plaquetas/citología , Plaquetas/efectos de los fármacos , Efrina-A4/agonistas , Efrina-A4/metabolismo , Humanos , Ligandos , Fosfoproteínas/agonistas , Fosfoproteínas/antagonistas & inhibidores , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Análisis por Matrices de Proteínas , Antagonistas del Receptor Purinérgico P2/farmacología , Receptor Cross-Talk , Receptor EphA4/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Vesículas Secretoras/efectos de los fármacos , Transducción de Señal
8.
PLoS One ; 6(11): e26238, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22073153

RESUMEN

BACKGROUND: We have shown that 1,2,3,4,6-penta-O-galloyl-α-D-glucopyranose (α-PGG), an orally effective hypoglycemic small molecule, binds to insulin receptors and activates insulin-mediated glucose transport. Insulin has been shown to bind to its receptors on platelets and inhibit platelet activation. In this study we tested our hypothesis that if insulin possesses anti-platelet properties then insulin mimetic small molecules should mimic antiplatelet actions of insulin. PRINCIPAL FINDINGS: Incubation of human platelets with insulin or α-PGG induced phosphorylation of insulin receptors and IRS-1 and blocked ADP or collagen induced aggregation. Pre-treatment of platelets with α-PGG inhibited thrombin-induced release of P-selectin, secretion of ATP and aggregation. Addition of ADP or thrombin to platelets significantly decreased the basal cyclic AMP levels. Pre-incubation of platelets with α-PGG blocked ADP or thrombin induced decrease in platelet cyclic AMP levels but did not alter the basal or PGE(1) induced increase in cAMP levels. Addition of α-PGG to platelets blocked agonist induced rise in platelet cytosolic calcium and phosphorylation of Akt. Administration of α-PGG (20 mg kg(-1)) to wild type mice blocked ex vivo platelet aggregation induced by ADP or collagen. CONCLUSIONS: These data suggest that α-PGG inhibits platelet activation, at least in part, by inducing phosphorylation of insulin receptors leading to inhibition of agonist induced: (a) decrease in cyclic AMP; (b) rise in cytosolic calcium; and (c) phosphorylation of Akt. These findings taken together with our earlier reports that α-PGG mimics insulin signaling suggest that inhibition of platelet activation by α-PGG mimics antiplatelet actions of insulin.


Asunto(s)
Taninos Hidrolizables/farmacología , Insulina/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Adenosina Trifosfato/metabolismo , AMP Cíclico/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Imitación Molecular , Selectina-P/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo
9.
J Mol Cell Cardiol ; 51(3): 347-56, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21651913

RESUMEN

Sudden cardiac death remains one of the most prevalent modes of death and is mainly caused by ventricular fibrillation (VF) in the setting of acute ischemia resulting from coronary thrombi. Animal experiments have shown that platelet activation may increase susceptibility of ischemic myocardium to VF, but the mechanism is unknown. In the present study, we evaluated the effects of activated blood platelet products (ABPPs) on electrophysiological properties and intracellular Ca(2+) (Ca(2+)(i)) homeostasis. Platelets were collected from healthy volunteers. After activation, their secreted ABPPs were added to superfusion solutions. Rabbit ventricular myocytes were freshly isolated, and membrane potentials and Ca(2+)(i) were recorded using patch-clamp methodology and indo-1 fluorescence measurements, respectively. ABPPs prolonged action potential duration and induced early and delayed afterdepolarizations. ABPPs increased L-type Ca(2+) current (I(Ca,L)) density, but left densities of sodium current, inward rectifier K(+) current, transient outward K(+) current, and rapid component of the delayed rectifier K(+) current unchanged. ABPPs did not affect kinetics or (in)activation properties of membrane currents. ABPPs increased systolic Ca(2+)(i), Ca(2+)(i) transient amplitude, and sarcoplasmic reticulum Ca(2+) content. ABPPs did not affect the Na(+)-Ca(2+) exchange current (I(NCX)) in Ca(2+)-buffered conditions. Products secreted from activated human platelets induce changes in I(Ca,L) and Ca(2+)(i), which result in action potential prolongation and the occurrence of early and delayed afterdepolarizations in rabbit myocytes. These changes may trigger and support reentrant arrhythmias in ischemia models of coronary thrombosis.


Asunto(s)
Plaquetas/metabolismo , Miocitos Cardíacos/fisiología , Activación Plaquetaria/fisiología , Función Ventricular , Potenciales de Acción/efectos de los fármacos , Animales , Factores Biológicos/metabolismo , Factores Biológicos/farmacología , Calcio/metabolismo , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Miocitos Cardíacos/efectos de los fármacos , Conejos , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Tripsina/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 31(1): 34-42, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21051668

RESUMEN

OBJECTIVE: Scavenger receptor BI (SR-BI) is a cell surface receptor that promotes the selective uptake of cholesteryl esters from high-density lipoprotein (HDL) by the liver. In mice, SR-BI deficiency results in increased plasma HDL cholesterol levels and enhanced susceptibility to atherosclerosis. The aim of this study was to investigate the role of SR-BI deficiency on platelet function. METHODS AND RESULTS: SR-BI-deficient mice were thrombocytopenic, and their platelets were abnormally large, probably because of an increased cholesterol content. The FeCl(3) acute injury model to study arterial thrombosis susceptibility showed that SR-BI wild-type mice developed total arterial occlusion after 24±2 minutes. In SR-BI-deficient mice, however, the time to occlusion was reduced to 13±1 minutes (P=0.02). Correspondingly, in SR-BI-deficient mice, platelets circulated in an activated state and showed increased adherence to immobilized fibrinogen. In contrast, platelet-specific disruption of SR-BI by bone marrow transplantation in wild-type mice did not alter plasma cholesterol levels or affect platelet count, size, cholesterol content, or reactivity, suggesting that changes in plasma cholesterol levels were responsible for the altered responsiveness of platelets in SR-BI-deficient mice. CONCLUSIONS: The function of SR-BI in HDL cholesterol homeostasis and prevention of atherosclerosis is indirectly also essential for maintaining normal platelet function and prevention of thrombosis.


Asunto(s)
Arteriopatías Oclusivas/metabolismo , Plaquetas/metabolismo , HDL-Colesterol/sangre , Activación Plaquetaria , Receptores Depuradores de Clase B/deficiencia , Trombosis/metabolismo , Animales , Arteriopatías Oclusivas/inducido químicamente , Arteriopatías Oclusivas/genética , Arteriopatías Oclusivas/patología , Arteriopatías Oclusivas/prevención & control , Plaquetas/patología , Trasplante de Médula Ósea , Cloruros , Colesterol en la Dieta/metabolismo , Modelos Animales de Enfermedad , Compuestos Férricos , Fibrinógeno/metabolismo , Ratones , Ratones Noqueados , Adhesividad Plaquetaria , Agregación Plaquetaria , Receptores Depuradores de Clase B/genética , Trombocitopenia/metabolismo , Trombocitopenia/patología , Trombosis/inducido químicamente , Trombosis/genética , Trombosis/patología , Trombosis/prevención & control , Factores de Tiempo , Regulación hacia Arriba
11.
Haematologica ; 95(12): 2005-12, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20713462

RESUMEN

BACKGROUND: The collagen receptor glycoprotein VI generates activating signals through an immunoreceptor tyrosine-based activating motif on the co-associated Fc receptor gamma chain. Leukocyte-associated immunoglobulin-like receptor-1 also ligates collagen but generates inhibitory signals through immunoreceptor tyrosine-based inhibitory motifs. Thus far, the cellular expression of glycoprotein VI and leukocyte-associated immunoglobulin-like receptor-1 appears mutually exclusive. DESIGN AND METHODS: Using flow cytometry, we studied expression of collagen receptors on differentiating human megakaryocytes. CD34(+) cells were isolated from umbilical cord blood and matured to megakaryocytes in vitro. Freshly isolated bone marrow cells were used to study primary megakaryocytes. Upon cell sorting, cytospins were made to examine cytological characteristics of differentiation. RESULTS: Megakaryocyte maturation is accompanied by up-regulation of glycoprotein VI and down-regulation of leukocyte-associated immunoglobulin-like receptor-1. Interestingly, both in cultures from hematopoietic stem cells and primary cells obtained directly from bone marrow, we identified a subset of morphologically distinct megakaryocytes which co-express glycoprotein VI and leukocyte-associated immunoglobulin-like receptor-1. CONCLUSIONS: This is the first report of a primary cell that co-expresses these collagen receptors with opposite signaling properties. Since megakaryocytes mature in the collagen-rich environment of the bone marrow, these findings may point to a role for leukocyte-associated immunoglobulin-like receptor-1 in the control of megakaryocyte maturation/migration.


Asunto(s)
Plaquetas/metabolismo , Células Progenitoras de Megacariocitos/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Inmunológicos/metabolismo , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Sangre Fetal/citología , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Integrina alfa2beta1/metabolismo , Megacariocitos/metabolismo , Células Madre Multipotentes/metabolismo , Receptores de Colágeno/metabolismo , Trombopoyesis
12.
Thromb Haemost ; 103(5): 1033-43, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20216992

RESUMEN

Incubation at 0 degrees C is known to expose b- N -acetyl-D-glucosamine residues on glycoprotein (GP) Ibalpha inducing receptor clustering and alpha(M)beta(2)-mediated platelet destruction by macrophages. Here we show that incubation at 0/37 degrees C (4 hours at 0 degrees C, followed by 1 hour at 37 degrees C to mimic cold-storage and post-transfusion conditions) triggers a conformational change in the N -terminal flank (NTF, amino acids, aa 1-35) but not in aa 36-282 of GPIbalpha as detected by antibody binding. Addition of the sugar N -acetyl-D-glucosamine (GN) inhibits responses induced by 0/37 degrees C. Incubation at 0 degrees C shifts GPIbalpha from the membrane skeleton to the cytoskeleton. Different GPIbalpha conformations have little effect on VWF/ristocetin-induced aggregation, but arrest of NTF change by GN interferes with agglutination and spreading on a VWF-coated surface under flow. Strikingly, incubation at 0/37 degrees C initiates thromboxane A(2) formation through a von Willebrand factor (VWF)-independent and GPIbalpha-dependent mechanism, as confirmed in VWF- and GPIbalpha-deficient platelets. We conclude that the NTF change induced by 0/37 degrees C incubation reflects clustering of GPIbalpha supports VWF/ristocetin-induced agglutination and spreading and is sufficient to initiate platelet activation in the absence of VWF.


Asunto(s)
Acetilglucosamina/metabolismo , Plaquetas/metabolismo , Conservación de la Sangre , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Enfermedades de von Willebrand/metabolismo , Acetilglucosamina/química , Anticuerpos Monoclonales , Plaquetas/patología , Células Cultivadas , Citoesqueleto/metabolismo , Humanos , Activación de Macrófagos , Antígeno de Macrófago-1/metabolismo , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Complejo GPIb-IX de Glicoproteína Plaquetaria/inmunología , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Agregación de Receptores/fisiología , Temperatura , Tromboxano A2/biosíntesis , Tromboxano A2/genética , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/patología , Enfermedades de von Willebrand/terapia , Factor de von Willebrand/metabolismo
13.
Diabetes ; 59(6): 1487-95, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20200314

RESUMEN

OBJECTIVE: Patients with type 2 diabetes have an increased risk of cardiovascular disease and show abnormalities in the coagulation cascade. We investigated whether increased synthesis of tissue factor (TF) by platelets could contribute to the hypercoagulant state. RESEARCH DESIGN AND METHODS: Platelets from type 2 diabetic patients and matched control subjects were adhered to different surface-coated proteins, and TF premRNA splicing, TF protein, and TF procoagulant activity were measured. RESULTS: Different adhesive proteins induced different levels of TF synthesis. A mimetic of active clopidogrel metabolite (AR-C69931 MX) reduced TF synthesis by 56 +/- 10%, an aspirin-like inhibitor (indomethacin) by 82 +/- 9%, and the combination by 96 +/- 2%, indicating that ADP release and thromboxane A(2) production followed by activation of P2Y12 and thromboxane receptors mediate surface-induced TF synthesis. Interference with intracellular pathways revealed inhibition by agents that raise cAMP and interfere with phosphatidylinositol 3-kinase/protein kinase B. Insulin is known to raise cAMP in platelets and inhibited collagen III-induced TF premRNA splicing and reduced TF activity by 35 +/- 5 and 47 +/- 5% at 1 and 100 nmol/l. Inhibition by insulin was reduced in type 2 diabetes platelets resulting in an approximately 1.6-fold higher TF synthesis than in matched control subjects. CONCLUSIONS: We characterized the extra- and intracellular mechanisms that couple surface activation to TF synthesis in adhering platelets. In healthy individuals, TF synthesis is inhibited by insulin, but in patients with type 2 diabetes inhibition is impaired. This leads to the novel finding that platelets from type 2 diabetic patients produce more TF than platelets from matched control subjects.


Asunto(s)
Plaquetas/fisiología , Diabetes Mellitus Tipo 2/sangre , Insulina/farmacología , Tromboplastina/biosíntesis , Fosfatasa Alcalina/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Presión Sanguínea , Diabetes Mellitus Tipo 2/fisiopatología , Factor X/efectos de los fármacos , Factor X/metabolismo , Hemoglobina Glucada/metabolismo , Humanos , Insulina/genética , Persona de Mediana Edad , Activación Plaquetaria , Agregación Plaquetaria/efectos de los fármacos , Precursores del ARN/genética , Empalme del ARN , Proteínas Recombinantes/farmacología , Tromboplastina/efectos de los fármacos , Tromboplastina/genética
14.
Blood ; 114(20): 4323-4, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19965704
15.
Am J Physiol Gastrointest Liver Physiol ; 296(4): G963-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19246633

RESUMEN

The serotonin transporter is implicated in the uptake of the vasoconstrictor serotonin from the circulation into the platelets, where 95% of all blood serotonin is stored and released in response to vascular injury. In vivo studies indicated that platelet-derived serotonin mediates liver regeneration after partial hepatectomy. We have recently generated serotonin transporter knockout rats and demonstrated that their platelets were almost completely depleted of serotonin. Here we show that these rats exhibit impaired hemostasis and contain about 1-6% of wild-type serotonin levels in the blood. Despite the marked reduction of serotonin levels in blood and platelets, efficient liver regeneration and collagen-induced platelet aggregation occur in rats lacking the serotonin transporter. These results provide evidence that liver regeneration is not dependent on the release of serotonin from platelets. Our findings indicate that very low levels of serotonin in blood are sufficient for liver regeneration.


Asunto(s)
Regeneración Hepática/genética , Regeneración Hepática/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Serotonina/farmacología , Animales , Tiempo de Sangría , Plaquetas/metabolismo , Eliminación de Gen , Hepatectomía , Homeostasis/genética , Homeostasis/fisiología , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 29(3): 372-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19096001

RESUMEN

OBJECTIVE: The sensitivity of platelets to aggregating agents increases when low-density lipoprotein (LDL) binds to apolipoprotein E receptor 2' (apoER2'), triggering activation of p38MAPK and formation of thromboxane A2. LDL signaling is terminated by PECAM-1 through recruitment and activation of the Ser/Thr protein phosphatase PP2A, but platelets remain unresponsive to LDL when PECAM-1 activation disappears. We report a second mechanism that halts LDL signaling and in addition lowers platelet responsiveness to aggregating agents. METHODS AND RESULTS: After a first stimulation with LDL, platelets remain unresponsive to LDL for 60 minutes, despite normal apoER2' activation by a second dose of LDL. A possible cause is persistent activation of the tyrosine phosphatases SHP-1 and SHP-2, which may not only block a second activation of p38MAPK, PECAM-1, and PP2A by LDL but also seem to reduce aggregation by TRAP, collagen, and ADP. CONCLUSION: These findings reveal that p38MAPK phosphorylation and platelet activation by LDL are suppressed by two mechanisms: (1) short activation of PECAM-1/PP2A, and (2) prolonged activation of SHP-1 and SHP-2. Activation of SHP-1 and SHP-2 is accompanied by reduced responsiveness to aggregating agents, which--if present in vivo--would make LDL an aggregation inhibitor during prolonged contact with platelets.


Asunto(s)
Plaquetas/enzimología , Lipoproteínas LDL/metabolismo , Agregación Plaquetaria , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Transducción de Señal , Adenosina Difosfato/metabolismo , Colágeno/metabolismo , Regulación hacia Abajo , Humanos , Proteínas Relacionadas con Receptor de LDL , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína Fosfatasa 2/metabolismo , Receptores de Lipoproteína/metabolismo , Receptores de Trombina/metabolismo , Tromboxano A2/metabolismo , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Thromb Haemost ; 99(6): 1068-78, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18521510

RESUMEN

Several studies have highlighted a specific role for membrane cholesterol domains in platelet signalling. Upon adhesion to von Willebrand factor (VWF) or collagen, cholesterol-rich domains (CRDs) accumulate in filopodial extensions and selectively harbour counterpart receptors (GPIb and GPVI) and associated signalling molecules. In the present study we have addressed the role of membrane cholesterol in Ca(2+) signalling and secretion during the interaction of platelets with VWF and collagen. VWF/ristocetin-induced platelet aggregation was delayed after treatment with methyl beta-cyclodextrin (mbCD), but the maximal aggregation response was not affected. Platelet spreading but not adhesion to immobilised VWF under flow was attenuated by cholesterol removal, and accompanied by moderate lowering in the spiking Ca(2+) response. On the other hand, platelet interaction with collagen was quite sensitive to cholesterol depletion. Platelet aggregation decreased after treatment with mbCD, and Ca(2+) responses were decreased, both under static and flow conditions. Cholesterol depletion affected the secondary feedback activation via release of thromboxane A(2) and ADP. The collagen-induced secretion of alpha granules and surface translocation of P-selectin and CD63 was also critically affected by cholesterol depletion. Confocal microscopy showed localization of p-Tyr at sites of contact with substrate and other platelets, where also CRDs accumulate. Our data thus reveal a more critical role for membrane cholesterol in collagen-induced than in VWF-induced Ca(2+) signalling, and furthermore support the concept that secondary activation responses are dependent on intact CRDs.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Membrana Celular/metabolismo , Colesterol/metabolismo , Colágeno Tipo III/metabolismo , Factor de von Willebrand/metabolismo , Adenosina Difosfato/metabolismo , Antígenos CD/metabolismo , Comunicación Autocrina , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Señalización del Calcio/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Colesterol/deficiencia , Hemorreología , Humanos , Microscopía Confocal , Selectina-P/metabolismo , Fosforilación , Adhesividad Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Transporte de Proteínas , Receptores de Colágeno/metabolismo , Vesículas Secretoras/metabolismo , Estrés Mecánico , Tetraspanina 30 , Tromboxano A2/metabolismo , Factores de Tiempo , Tirosina/metabolismo , beta-Ciclodextrinas/farmacología
18.
Int J Biochem Cell Biol ; 40(11): 2374-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18468940

RESUMEN

There is a strong correlation between the level of plasma low-density lipoprotein (LDL) and death by cardiovascular disease (CVD). As a main carrier of cholesterol, a high low-density lipoprotein concentration stimulates atherogenesis by its capacity to become oxidized and to become endocytosed by macrophages in the vessel wall forming cholesterol-rich plaques that are sites for arterial occlusion. New evidence points at a second role of low-density lipoprotein in increasing cardiovascular disease-risk. Contact with low-density lipoprotein induces platelet hypersensitivity to agonists that initiate platelet functions thereby enhancing adhesion, aggregation and secretion of granule contents. The signalling pathways that mediate the priming of platelets by native and oxidized low-density lipoprotein have now been characterized.


Asunto(s)
Plaquetas/metabolismo , Lipoproteínas LDL/metabolismo , Activación Plaquetaria , Transducción de Señal/fisiología , Humanos , Lipoproteínas LDL/química , Oxidación-Reducción , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 27(11): 2476-83, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17761940

RESUMEN

OBJECTIVE: The interaction of platelets with low density lipoprotein (LDL) contributes to the development of cardiovascular disease. Platelets are activated by native LDL (nLDL) through apoE Receptor 2' (apoER2')-mediated signaling to p38(MAPK) and by oxidized LDL (oxLDL) through lysophosphatidic acid (LPA) signaling to Rho A and Ca2+. Here we report a new mechanism for platelet activation by oxLDL. METHODS AND RESULTS: Oxidation of nLDL increases p38(MAPK) activation through a mechanism that is (1) independent of LPA, and (2) unlike nLDL-signaling not desensitized by prolonged platelet-LDL contact or inhibited by receptor-associated protein or chondroitinase ABC. Antibodies against scavenger receptors CD36 and SR-A alone fail to block p38(MAPK) activation by oxLDL but combined blockade inhibits p38(MAPK) by >40% and platelet adhesion to fibrinogen under flow by >60%. Mouse platelets deficient in either CD36 or SR-A show normal p38(MAPK) activation by oxLDL but combined deficiency of CD36 and SR-A disrupts oxLDL-induced activation of p38(MAPK) by >70%. CONCLUSION: These findings reveal a novel platelet-activating pathway stimulated by oxLDL that is initiated by the combined action of CD36 and SR-A.


Asunto(s)
Antígenos CD36/fisiología , Lipoproteínas LDL/fisiología , Activación Plaquetaria/fisiología , Receptores Depuradores de Clase A/fisiología , Animales , Plaquetas , Humanos , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Clin Cancer Res ; 13(18 Pt 1): 5341-7, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17855648

RESUMEN

PURPOSE: One of the key factors that promotes angiogenesis is vascular endothelial growth factor (VEGF). Platelets are the main source of VEGF in blood and contribute to angiogenesis by release of growth factors, including VEGF, from their alpha-granules on activation. The monoclonal antibody bevacizumab blocks VEGF in the blood of patients within hours after administration. Platelets are known to endocytose plasma proteins including immunoglobulins. We tested the hypothesis that platelets take up bevacizumab. EXPERIMENTAL DESIGN: Fluorescence-activated cell sorting analysis, immunofluorescence imaging, and Western blotting were used to study uptake and release of bevacizumab by platelets in vitro and in vivo. The angiogenic activity of platelets preincubated with bevacizumab was studied in endothelial proliferation assays. Finally, we determined whether treatment with bevacizumab neutralizes VEGF in platelets from cancer patients. RESULTS: We found that platelets are able to take up bevacizumab. Activation of platelets preincubated with bevacizumab resulted in release of the antibody and release of VEGF neutralized by bevacizumab. Immunofluorescence microscopy revealed that FITC-labeled bevacizumab and P-selectin colocalize, indicating alpha-granule localization. In addition, bevacizumab uptake inhibited platelet-induced human endothelial cell proliferation. In in vivo rabbit experiments, FITC-labeled bevacizumab was present in platelets after 2 h and up to 2 weeks following i.v. administration. Finally, we found that platelets take up bevacizumab in patients receiving bevacizumab treatment. Within 8 h after bevacizumab administration, platelet VEGF was almost completely neutralized due to this uptake. CONCLUSION: These studies show that bevacizumab is taken up by platelets and may explain its clinical effect on wound healing and tumor growth.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Plaquetas/metabolismo , Neovascularización Fisiológica , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales Humanizados , Bevacizumab , Plaquetas/química , Citometría de Flujo , Humanos , Transporte de Proteínas , Conejos , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...