Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Blood ; 114(7): 1405-16, 2009 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-19429868

RESUMEN

Platelet response to activation varies widely between individuals but shows interindividual consistency and strong heritability. The genetic basis of this variation has not been properly explored. We therefore systematically measured the effect on function of sequence variation in 97 candidate genes in the collagen and adenosine-diphosphate (ADP) signaling pathways. Resequencing of the genes in 48 European DNA samples nearly doubled the number of known single nucleotide polymorphisms (SNPs) and informed the selection of 1327 SNPs for genotyping in 500 healthy Northern European subjects with known platelet responses to collagen-related peptide (CRP-XL) and ADP. This identified 17 novel associations with platelet function (P < .005) accounting for approximately 46% of the variation in response. Further investigations with platelets of known genotype explored the mechanisms behind some of the associations. SNPs in PEAR1 associated with increased platelet response to CRP-XL and increased PEAR1 protein expression after platelet degranulation. The minor allele of a 3' untranslated region (UTR) SNP (rs2769668) in VAV3 was associated with higher protein expression (P = .03) and increased P-selectin exposure after ADP activation (P = .004). Furthermore the minor allele of the intronic SNP rs17786144 in ITPR1 modified Ca(2+) levels after activation with ADP (P < .004). These data provide novel insights into key hubs within platelet signaling networks.


Asunto(s)
Plaquetas/fisiología , Degranulación de la Célula/genética , Regulación de la Expresión Génica/fisiología , Activación Plaquetaria/genética , Sitios de Carácter Cuantitativo/fisiología , Transducción de Señal/genética , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/metabolismo , Adenosina Difosfato/genética , Adenosina Difosfato/metabolismo , Alelos , Plaquetas/citología , Colágeno/genética , Colágeno/metabolismo , Europa (Continente) , Femenino , Genómica , Genotipo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/biosíntesis , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Selectina-P/genética , Selectina-P/metabolismo , Polimorfismo de Nucleótido Simple , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Población Blanca
8.
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
9.
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
10.
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
11.
Alcohol Clin Exp Res ; 31(8): 1308-14, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17559545

RESUMEN

BACKGROUND: Moderate and prolonged consumption of red wine is associated with decreased cardiovascular morbidity and mortality. Inhibition of platelet functions by ingredients in red wine is thought to be one of the causes. However, the molecular mechanism of this inhibition has remained unexplained. MATERIALS AND METHODS: We measured aggregation, changes in cytosolic Ca(2+) and tyrosine phosphorylation of the inhibitory receptor platelet endothelial cell adhesion molecule-1 (PECAM-1) in platelets stimulated with thrombin receptor (PAR-1) activating peptide (TRAP) and ADP and investigated the effects of alcohol-free polyphenolic grape extract (PGE), alcohol, and the polyphenols catechin, epi-catechin, resveratrol, trans-resveratrol, and gallic acid. RESULTS: Polyphenolic grape extract induced dose-dependent inhibition of TRAP-induced and ADP-induced platelet aggregation and Ca(2+) mobilization. Inhibition was accompanied by activation of PECAM-1. Apart from a slight inhibition by catechin, ethanol or other individual polyphenols failed to inhibit aggregation or activate PECAM-1. CONCLUSIONS: Red wine inhibits platelet functions through its PGE content, which stimulates the inhibitory receptor PECAM-1, thereby attenuating platelet activation.


Asunto(s)
Enfermedades Cardiovasculares/epidemiología , Flavonoides/farmacología , Fenoles/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Vitis/química , Vino , Adenosina Difosfato/farmacología , Biotransformación/efectos de los fármacos , Calcio/metabolismo , Citosol/metabolismo , Flavonoides/aislamiento & purificación , Francia/epidemiología , Frutas/química , Humanos , Indicadores y Reactivos , Fenoles/aislamiento & purificación , Fosforilación , Extractos Vegetales/química , Extractos Vegetales/farmacología , Agregación Plaquetaria/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Polifenoles , Receptores de Trombina/metabolismo , Tirosina/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 27(7): 1657-65, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17510465

RESUMEN

OBJECTIVE: Protein misfolding diseases result from the deposition of insoluble protein aggregates that often contain fibrils called amyloid. Amyloids are found in Alzheimer disease, atherosclerosis, diabetes mellitus, and systemic amyloidosis, which are diseases where platelet activation might be implicated. METHODS AND RESULTS: We induced amyloid properties in 6 unrelated proteins and found that all induced platelet aggregation in contrast to fresh controls. Amyloid-induced platelet aggregation was independent of thromboxane A2 formation and ADP secretion but enhanced by feedback stimulation through these pathways. Treatments that raised cAMP (iloprost), sequestered Ca2+ (BAPTA-AM) or prevented amyloid-platelet interaction (sRAGE, tissue-type plasminogen activator [tPA]) induced almost complete inhibition. Modulation of the function of CD36 (CD36-/- mice), p38(MAPK) (SB203580), COX-1 (indomethacin), and glycoprotein Ib alpha (Nk-protease, 6D1 antibody) induced approximately 50% inhibition. Interference with fibrinogen binding (RGDS) revealed a major contribution of alphaIIb beta3-independent aggregation (agglutination). CONCLUSIONS: Protein misfolding resulting in the appearance of amyloid induces platelet aggregation. Amyloid activates platelets through 2 pathways: one is through CD36, p38(MAPK), thromboxane A2-mediated induction of aggregation; the other is through glycoprotein Ib alpha-mediated aggregation and agglutination. The platelet stimulating properties of amyloid might explain the enhanced platelet activation observed in many diseases accompanied by the appearance of misfolded proteins with amyloid.


Asunto(s)
Amiloide/farmacología , Plaquetas/citología , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/farmacología , Plaquetas/metabolismo , Antígenos CD36/metabolismo , Células Cultivadas , Humanos , Agregación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Valores de Referencia , Sensibilidad y Especificidad , Tromboxano A2/metabolismo , Activador de Tejido Plasminógeno/metabolismo
13.
Transfusion ; 46(12): 2090-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17176320

RESUMEN

BACKGROUND: Platelet (PLT) storage at 0 to 4 degrees C suppresses bacterial multiplication, but induces clusters of glycoprotein (GP) Ibalpha that trigger their phagocytosis by macrophages and reduce their survival after transfusion. A method was sought that detects cold-induced changes in GPIbalpha involved in phagocytosis. STUDY DESIGN AND METHODS: Human PLTs were isolated and stored for up to 48 hours at 0 degrees C. Binding of a phycoerythrin (PE)-labeled antibody directed against amino acids (AA) 1-35 on GPIbalpha (AN51-PE) was compared with phagocytosis of PLTs by matured monocytic THP-1 cells, analyzed by fluorescence-activated cell sorting. RESULTS: Freshly isolated PLTs were detected as a single population of AN51-PE-positive particles and showed less than 5 percent phagocytosis. Cold storage led to a decrease in AN51-PE binding and an increase in phagocytosis. N-Acetylglucosamine, known to interfere with macrophage recognition of GPIbalpha clusters, restored normal AN51-PE binding to cold-stored PLTs and suppressed phagocytosis. CONCLUSIONS: It is concluded that binding of an antibody against AA 1-35 on GPIbalpha reflects changes in GPIbalpha that make PLTs targets for phagocytosis by macrophages.


Asunto(s)
Plaquetas/fisiología , Macrófagos/inmunología , Fagocitosis , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Acetilglucosamina/farmacología , Anticuerpos Monoclonales/metabolismo , Filtración , Humanos
14.
Transfusion ; 46(8): 1432-43, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16934082

RESUMEN

BACKGROUND: Earlier it was reported that metabolic arrest followed by incubation at 4 degrees C reduces the platelet (PLT) storage defect. Here it is reported that this treatment also reduces binding and phagocytosis by macrophages. STUDY DESIGN AND METHODS: Phagocytosis of mepacrine-labeled PLTs by macrophages changes the latter into bright fluorescent particles easily detected by fluorescence-activated cell sorting. RESULTS: In combination with conventional binding analysis it was found that binding to phorbol 12-myristate 13-acetate-matured THP-1 cells is primarily regulated by PLT P-selectin expression and phagocytosis by combined phosphatidylserine (PS) exposure and glycoprotein (GP) Ibalpha clustering. It was found that trapping of PLT Ca2+ and raising cAMP reduces phagocytosis by lowering PS exposure. Chilling of PLTs leads to an increase in binding and PS- and GPIbalpha-mediated phagocytosis. Prior depletion of PLT energy stores prevents this increase by preserving low Ca2+ concentration, PS exposure, and PS-mediated phagocytosis. CONCLUSION: These data characterize the individual factors that control PLT binding and phagocytosis and might help to define conditions that improve the survival of stored PLTs after transfusion.


Asunto(s)
Conservación de la Sangre , Macrófagos , Fagocitosis , Adhesividad Plaquetaria , Conservación de la Sangre/efectos adversos , Línea Celular , Supervivencia Celular , Humanos , Macrófagos/metabolismo , Selectina-P/metabolismo , Transfusión de Plaquetas
15.
Transfusion ; 46(6): 1018-28, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16734820

RESUMEN

BACKGROUND: Platelets (PLTs) contain purinergic receptors for ATP (P2X1) and ADP (P2Y1 and P2Y12) that rapidly desensitize upon stimulation with these nucleotides. In vivo, this is antagonized by ectonucleotidases on the surface of endothelial cells and white blood cells (WBCs). The receptor desensitization of ATP- and ADP-induced responses of PLTs stored in plasma without WBCs was investigated. STUDY DESIGN AND METHODS: ATP- and ADP-induced PLT shape change (shear-induced) aggregation and Ca2+ signaling were measured in the presence or absence of plasma. Degradation of nucleotides in plasma was quantified by high-performance liquid chromatography. RESULTS: Washed PLTs became refractory for ATP and ADP in shape change, aggregation, and Ca2+ responses during a 90-minute incubation at 37 degrees C. The PLT responses mediated by P2X1, P2Y1, and P2Y12 receptors gradually reduced or disappeared. When plasma was present, however, the PLTs persistently showed high responses to ATP and ADP. Heat treatment of plasma abolished this effect. Also under conditions of flow and high shear, PLTs in plasma kept high P2X1 activity, mediating aggregate formation. In isolated plasma, not containing WBCs, nucleotides were degraded in the order of ADP/UDP>ATP/UTP. Degradation of ATP was partly inhibited by blocking the ecto-NTPDase CD39, whereas degradation of both ATP and ADP was inhibited by blocking ectopyrophosphatase/phosphodiesterase activity. Part of the nucleotide-degrading activities appeared to be membrane-bound. CONCLUSION: Ectonucleotidases in plasma preserve the functionality of P2X1 and P2Y receptors. Upon PLT storage, these plasma activities are essential to ensure adequate (shear-dependent) formation of aggregates and thrombi.


Asunto(s)
Plaquetas/fisiología , Conservación de la Sangre , Pirofosfatasas/fisiología , Receptores Purinérgicos/metabolismo , Trombosis/etiología , Adenosina Difosfato/farmacología , Adenosina Trifosfato/farmacología , Señalización del Calcio , Humanos , Plasma/enzimología , Activación Plaquetaria , Pirofosfatasas/sangre , Receptores Purinérgicos P2 , Receptores Purinérgicos P2X
16.
Arterioscler Thromb Vasc Biol ; 26(2): 417-22, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16339499

RESUMEN

OBJECTIVE: ADP-induced P2y12 signaling is crucial for formation and stabilization of an arterial thrombus. We demonstrated recently in platelets from healthy subjects that insulin interferes with Ca2+ increases induced by ADP-P2y1 contact through blockade of the G-protein Gi, and thereby with P2y12-mediated suppression of cAMP. METHODS AND RESULTS: Here we show in patients with type 2 diabetes mellitus (DM2) that platelets have lost responsiveness to insulin leading to increased adhesion, aggregation, and procoagulant activity on contact with collagen. Using Ser473 phosphorylation of protein kinase B as output for insulin signaling, a 2-fold increase is found in insulin-stimulated normal platelets, but in DM platelets there is no significant response. In addition, DM2 platelets show increased P2y12-mediated suppression of cAMP and decreased P2y12 inhibition by the receptor antagonist AR-C69931MX. CONCLUSIONS: The loss of responsiveness to insulin together with increased signaling through P2y12 might explain the hyperactivity of platelets in patients with DM2.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Diabetes Mellitus Tipo 2/sangre , Hipoglucemiantes/farmacología , Insulina/farmacología , Adenosina Difosfato/farmacología , Calcio/metabolismo , Colágeno/farmacología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Homeostasis , Humanos , Hipoglucemiantes/metabolismo , Técnicas In Vitro , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Proteínas de la Membrana/metabolismo , Perfusión , Fosfoproteínas/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Receptor de Insulina/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y12 , Transducción de Señal/efectos de los fármacos
17.
Arterioscler Thromb Vasc Biol ; 25(4): 867-72, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15692097

RESUMEN

OBJECTIVE: Because of the large variation in oxidizing procedures and susceptibility to oxidation of low-density lipoprotein (LDL) and the lack in quantification of LDL oxidation, the role of oxidation in LDL-platelet contact has remained elusive. This study aims to compare platelet activation by native LDL (nLDL) and oxidized LDL (oxLDL). METHODS AND RESULTS: After isolation, nLDL was dialyzed against FeSO4 to obtain LDL oxidized to well-defined extents varying between 0% and >60%. The oxLDL preparations were characterized with respect to their platelet-activating properties. An increase in LDL oxidation enhances platelet activation via 2 independent pathways, 1 signaling via p38(MAPK) phosphorylation and 1 via Ca2+ mobilization. Between 0% and 15% oxidation, the p38(MAPK) route enhances fibrinogen binding induced by thrombin receptor (PAR-1)-activating peptide (TRAP), and signaling via Ca2+ is absent. At >30% oxidation, p38(MAPK) signaling increases further and is accompanied by Ca2+ mobilization and platelet aggregation in the absence of a second agonist. Despite the increase in p38(MAPK) signaling, synergism with TRAP disappears and oxLDL becomes an inhibitor of fibrinogen binding. Inhibition is accompanied by binding of oxLDL to the scavenger receptor CD36, which is associated with the fibrinogen receptor, alpha(IIb)beta3. CONCLUSIONS: At >30% oxidation, LDL interferes with ligand binding to integrin alpha(IIb)beta3, thereby attenuating platelet functions.


Asunto(s)
Plaquetas/metabolismo , Lipoproteínas LDL/metabolismo , Agregación Plaquetaria/fisiología , Antígenos CD36/metabolismo , Señalización del Calcio/fisiología , AMP Cíclico/metabolismo , Humanos , Técnicas In Vitro , Cadenas alfa de Integrinas/metabolismo , Cadenas beta de Integrinas/metabolismo , Ligandos , Sistema de Señalización de MAP Quinasas/fisiología , Oxidación-Reducción , Unión Proteica/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Alcohol Clin Exp Res ; 28(10): 1562-8, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15597090

RESUMEN

BACKGROUND: Moderate alcohol consumption is associated with decreased mortality from cardiovascular disease. Drinking large amounts in a short period (binge drinking) is associated with increased cardiovascular morbidity. We tested whether rapid consumption of a large dose of alcohol affects platelet aggregation and adhesion. METHODS: Healthy volunteers (n = 20) were asked to drink three glasses of alcohol or red wine in a 45-min period. Thereafter, another 45 min was allowed for absorption of alcohol. Ninety minutes after the start of the experiment, blood was collected. This entire cycle was repeated once, resulting in consumption of six alcohol-containing drinks in 3 hr. Adenosine-diphosphate (ADP)-induced aggregation was measured and platelet adhesion to fibrinogen and collagen was measured in a perfusion chamber at shear rates of 300/sec and 1600/sec. Platelet coverage and aggregate size were measured. RESULTS: Acute alcohol intake significantly increased platelet aggregation in suspension when stimulated with low concentrations of ADP (0.1 and 0.5 microg/ml). This effect was not observed when consuming red wine. In contrast, adhesion to fibrinogen was significantly inhibited by alcohol but not red wine at high shear rate after six drinks (p = 0.025). The inhibition was accompanied by a reduction in aggregate size at 90 and 180 min after the start of the experiment. Adhesion to collagen was not altered by either alcohol or red wine. CONCLUSIONS: Rapid intake of alcohol increases platelet aggregation, which might contribute to the increased mortality associated with binge drinking. Red wine does not show increased platelet aggregation, which might support the reduction of cardiovascular disease in red wine consumers. However, alcohol inhibits platelet adhesion to fibrinogen-coated surface under flow. The diminished adhesion might contribute to the cardioprotective effects of alcohol.


Asunto(s)
Consumo de Bebidas Alcohólicas/sangre , Etanol/administración & dosificación , Fibrinógeno/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Adulto , Consumo de Bebidas Alcohólicas/efectos adversos , Análisis de Varianza , Femenino , Humanos , Masculino , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Factores de Tiempo , Vino
19.
J Histochem Cytochem ; 52(10): 1277-85, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15385574

RESUMEN

Localization and coordinate phosphorylation/dephosphorylation of endothelial nitric oxide synthase (eNOS) are critical determinants for the basal and stimulated production of nitric oxide. Several phosphorylation sites in eNOS have been identified as targets of the cAMP-dependent protein kinase A (PKA). Basal eNOS activity is also regulated by interaction with caveolin-1, the major coat protein of caveolae. In the present study we have examined in rat aorta endothelium the subcellular steady-state distribution of eNOS, the catalytic subunit of PKA (PKA-c), and caveolin-1. Basal eNOS expression was found in two distinct locations, the endothelial cell surface and the Golgi complex. Cell surface eNOS was equally distributed over caveolar and non-caveolar membranes but was 2.5-fold enriched on luminal lamellipodia located at endothelial cell contacts. PKA-c colocalized with eNOS in the lamellipodia, whereas caveolin-1 was absent from these membrane domains. PKA-c was also found associated with cell surface caveolae and with tubulovesicular membranes of Golgi complex and endosomes. The topological proximity of eNOS with the catalytic subunit of PKA in restricted intracellular locations may provide mechanisms for differential PKA-mediated eNOS regulation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endotelio Vascular/enzimología , Uniones Intercelulares/enzimología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/biosíntesis , Animales , Aorta/enzimología , Dominio Catalítico , Caveolas/enzimología , Caveolina 1 , Caveolinas/metabolismo , Células Cultivadas , Endotelio Vascular/ultraestructura , Técnica del Anticuerpo Fluorescente , Aparato de Golgi/enzimología , Microscopía Inmunoelectrónica , Óxido Nítrico Sintasa de Tipo III , Ratas
20.
J Biol Chem ; 279(5): 3254-64, 2004 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-14602724

RESUMEN

Platelet agonists initiate aggregation and secretion by activating receptors coupled to the G-protein G(q), thereby raising cytosolic Ca(2+), [Ca(2+)](i). The rise in [Ca(2+)](i) is facilitated via inhibition of cAMP formation by the inhibitory G-protein of adenylyl cyclase, G(i). Since insulin attenuates platelet activation, we investigated whether insulin interferes with cAMP regulation. Here we report that insulin (0.5-200 nmol/liter) interferes with agonist-induced increases in [Ca(2+)](i) (ADP, thrombin), cAMP suppression (thrombin), and aggregation (ADP). The effects of insulin are as follows: (i) independent of the P2Y(12) receptor, which mediates ADP-induced cAMP lowering; (ii) not observed during G(s)-mediated cAMP formation; (iii) unaffected by treatments that affect phosphodiesterases (3-isobutyl-1-methylxanthine); and (iv) not changed by interfering with NO-mediated regulation of cAMP degradation (N(G)-monomethyl-l-arginine). Hence, insulin might interfere with G(i). Indeed, insulin induces the following: (i) tyrosine phosphorylation of the insulin receptor, the insulin receptor substrate-1 (IRS-1) and G(i)alpha(2); (ii) co-precipitation of IRS-1 with G(i)alpha(2) but not with other G alpha subunits. Despite persistent receptor activation, the association of IRS-1 with G(i)alpha(2) is transient, being optimal at 5 min and 1 nmol/liter insulin, which is sufficient to suppress Ca(2+) signaling by ADP, and at 10 min and 100 nmol/liter insulin, which is required to suppress Ca(2+) signaling by thrombin. Epinephrine, a known platelet sensitizer and antagonist of insulin, abolishes the effect of insulin on [Ca(2+)](i), tyrosine phosphorylation of G(i)alpha(2), and aggregation by interfering with the phosphorylation of the insulin receptor beta subunit. We conclude that insulin attenuates platelet functions by interfering with cAMP suppression through IRS-1 and G(i).


Asunto(s)
Calcio/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Insulina/metabolismo , Fosfoproteínas/fisiología , Adenosina Difosfato/química , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Epinefrina/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina , Óxido Nítrico/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Activación Plaquetaria , Agregación Plaquetaria , Pruebas de Precipitina , Receptor de Insulina/metabolismo , Transducción de Señal , Trombina/farmacología , Factores de Tiempo , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA