Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Neurosci ; 13: 32, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30886568

RESUMO

Cigarette smoking is one of the major causes of coronary heart disease with a thirty percent mortality rate in the United States. Cigarette smoking acting on the central nervous system (CNS) to stimulate the sympathetic nervous system (SNS) through, which facilitates the secretion of serotonin (5-HT) and catecholamines to supraphysiological levels in blood. The enhanced levels of 5-HT and catecholamines in smokers' blood are associated with increases in G protein-coupled receptor signaling and serotonylation of small GTPases, which in turn lead to remodeling of cytoskeletal elements to enhance granule secretion and promote unique expression of sialylated N-glycan structures on smokers' platelets. These mechanisms enhance aggregation and adhesion of smokers' platelets relative to those of non-smokers. This review focuses on the known mechanisms by which 5-HT and SERT, in coordinated signaling with catecholamines, impacts cigarette smokers' platelet biology.

2.
J Am Heart Assoc ; 6(5)2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28522678

RESUMO

BACKGROUND: Cigarette smoking plays a major role in cardiovascular diseases. The acute effects of cigarette smoking produce central nervous system-mediated activation of the sympathetic nervous system. The overactive sympathetic nervous system stimulates the secretion of serotonin (5-HT) and catecholamine into blood at supraphysiological levels. The correlation between these pathological conditions induced by smoking and the increased risk of thrombosis has not been thoroughly investigated. The goal of our study was to explore cigarette smoking-associated changes in platelet biology mediated by elevated 5-HT and catecholamine levels in blood plasma. METHODS AND RESULTS: Using blood samples collected from healthy nonsmokers and smokers (15 minutes after smoking), we determined that cigarette smoking increased the plasma 5-HT/catecholamine concentration by several fold and the percent aggregation of platelets 2-fold. Liquid chromatography-tandem mass spectrometry analysis of proteins eluted from platelet plasma membranes of smokers and nonsmokers demonstrated that GTPase-activating proteins and proteins participating in the actin cytoskeletal network were differentially and significantly elevated in smokers' platelet membranes compared with those of nonsmokers. Interestingly, Matrix-assisted laser desorption/ionization-mass spectrometry analyses of the glycans eluted from platelet plasma membranes of the smokers demonstrated that the level and structures of glycans are different from the nonsmokers' platelet surface glycans. Pharmacological blockade of 5-HT or catecholamine receptors counteracted the 5-HT/catecholamine-mediated aggregation and altered the level and composition of glycan on platelet surfaces. CONCLUSIONS: Based on our findings, we propose that smoking-associated 5-HT/catecholamine signaling accelerates the trafficking dynamics of platelets, and this remodels the surface proteins and glycans and predisposes platelets to hyperactive levels. Smokers' platelets also had correspondingly higher resting concentrations of intracellular calcium and transglutaminase activity. These findings suggest a link among smoking, platelet 5-HT, catecholamine signaling, and their downstream effectors-including phospholipase C and inositol-1,4,5-triphosphate pathways-resulting in an increased tonic level of platelet activation in smokers.


Assuntos
Plaquetas/metabolismo , Membrana Celular/metabolismo , Epinefrina/sangue , Ativação Plaquetária , Serotonina/sangue , Transdução de Sinais , Fumar/sangue , Antagonistas Adrenérgicos beta/farmacologia , Adulto , Plaquetas/efeitos dos fármacos , Cálcio/sangue , Estudos de Casos e Controles , Membrana Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Humanos , Masculino , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Polissacarídeos/sangue , Transporte Proteico , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fumar/efeitos adversos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Fatores de Tempo , Transglutaminases/sangue , Regulação para Cima
3.
Blood ; 124(16): 2475-6, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25323685

RESUMO

In this issue of Blood, Darbousset et al define opposing roles for adenosine triphosphate (ATP) and adenosine in regulating polymorphonuclear neutrophil (PMN) activation, fibrin formation, and thrombus growth following vascular injury.


Assuntos
Plaquetas/metabolismo , Neutrófilos/metabolismo , Receptores Purinérgicos P2X1/genética , Trombose/genética , Animais , Humanos
4.
Blood ; 124(13): 2094-103, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-24948658

RESUMO

In vivo visualization of thrombopoiesis suggests an important role for shear flow in platelet biogenesis. In vitro, shear stress was shown to accelerate proplatelet formation from mature megakaryocytes (Mks). Yet, the role of biomechanical forces on Mk biology and platelet biogenesis remains largely unexplored. In this study, we investigated the impact of shear stress on Mk maturation and formation of platelet-like particles (PLPs), pro/preplatelets (PPTs), and Mk microparticles (MkMPs), and furthermore, we explored a physiological role for MkMPs. We found that shear accelerated DNA synthesis of immature Mks in an exposure time- and shear stress level-dependent manner. Both phosphatidylserine exposure and caspase-3 activation were enhanced by shear stress. Exposure to physiological shear dramatically increased generation of PLPs/PPTs and MkMPs by up to 10.8 and 47-fold, respectively. Caspase-3 inhibition reduced shear-induced PLP/PPT and MkMP formation. PLPs generated under shear flow displayed improved functionality as assessed by CD62P exposure and fibrinogen binding. Significantly, coculture of MkMPs with hematopoietic stem and progenitor cells promoted hematopoietic stem and progenitor cell differentiation to mature Mks synthesizing α- and dense-granules, and forming PPTs without exogenous thrombopoietin, thus identifying a novel and unexplored potential physiological role for MkMPs.


Assuntos
Diferenciação Celular , Micropartículas Derivadas de Células/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Resistência ao Cisalhamento , Estresse Fisiológico , Trombopoese/fisiologia , Antígenos CD34/metabolismo , Caspase 3/metabolismo , Micropartículas Derivadas de Células/ultraestrutura , Replicação do DNA , Ativação Enzimática , Humanos , Poliploidia
5.
Mol Pharmacol ; 86(1): 1-11, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24723492

RESUMO

It is now well accepted that protease activated receptor (PAR) 1 and PAR4 have differential roles in platelet activation. PAR4, a low-affinity thrombin receptor in human platelets, participates in sustained platelet activation in a P2Y12-dependent manner; however, the mechanisms are not defined. Our previous studies demonstrated that thrombin induces the association of PAR4 with P2Y12, together with arrestin recruitment to the complex. Here we show that PAR4 and P2Y12 directly interact to coregulate Akt signaling after PAR4 activation. We observed direct and specific interaction of P2Y12 with PAR4 but not PAR1 by bioluminescent resonance energy transfer when the receptors were coexpressed in human embryonic kidney 293T cells. PAR4-P2Y12 dimerization was promoted by PAR4-AP and inhibited by P2Y12 antagonist. By using sequence comparison of the transmembrane domains of PAR1 and PAR4, we designed a mutant form of PAR4, "PAR4SFT," by replacing LGL194-196 at the base of transmembrane domain 4 with the corresponding aligned PAR1 residues SFT 220-222. PAR4SFT supported only 8.74% of PAR4-P2Y12 interaction, abolishing P2Y12-dependent arrestin recruitment to PAR4 and Akt activation. Nonetheless, PAR4SFT still supported homodimerization with PAR4. PAR4SFT failed to induce a calcium flux when expressed independently; however, coexpression of increasing concentrations of PAR4SFT, together with PAR4 potentiated PAR4-mediated calcium flux, suggested that PAR4 act as homodimers to signal to Gq-coupled calcium responses. In conclusion, PAR4 LGL (194-196) governs agonist-dependent association of PAR4 with P2Y12 and contributes to Gq-coupled calcium responses. PAR4-P2Y12 association supports arrestin-mediated sustained signaling to Akt. Hence, PAR4-P2Y12 dimerization is likely to be important for the PAR4-P2Y12 dependent stabilization of platelet thrombi.


Assuntos
Arrestinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Receptores de Trombina/metabolismo , Cálcio/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Ativação Plaquetária/fisiologia , Estrutura Terciária de Proteína , Transdução de Sinais/fisiologia , Trombina/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 32(5): 1211-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22362759

RESUMO

OBJECTIVE: Members of the protein kinase C (PKC) family are shown to positively and negatively regulate platelet activation. Although positive regulatory roles are extensively studied, negative regulatory roles of PKCs are poorly understood. We investigated the mechanism and specific isoforms involved in PKC-mediated negative regulation of ADP-induced functional responses. METHODS AND RESULTS: A pan-PKC inhibitor, GF109203X, potentiated ADP-induced cPLA(2) phosphorylation and thromboxane generation as well as ERK activation and intracellular calcium (Ca(2+)(i)) mobilization, 2 signaling molecules, upstream of cPLA(2) activation. Thus, PKCs inhibit cPLA(2) activation by inhibiting ERK and Ca(2+)(i) mobilization. Because the inhibitor of classic PKC isoforms, GO-6976, did not affect ADP-mediated thromboxane generation, we investigated the role of novel class of PKC isoforms. ADP-induced thromboxane generation, calcium mobilization, and ERK phosphorylation were potentiated in PKCε null murine platelets compared with platelets from wild-type littermates. Interestingly, when thromboxane release is blocked, ADP-induced aggregation in PKCε knockout and wild-type was similar, suggesting that PKCε does not affect ADP-induced aggregation directly. PKCε knockout mice exhibited shorter times to occlusion in an FeCl(3)-induced arterial injury model and shorter bleeding times in tail-bleeding experiments. CONCLUSIONS: We conclude that PKCε negatively regulates ADP-induced thromboxane generation in platelets and offers protection against thrombosis.


Assuntos
Difosfato de Adenosina/farmacologia , Plaquetas/metabolismo , Cálcio/metabolismo , Agregação Plaquetária/fisiologia , Proteína Quinase C-épsilon/metabolismo , Tromboxanos/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Líquido Intracelular/metabolismo , Camundongos , Agregação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Transdução de Sinais , Trombose/metabolismo , Trombose/patologia
7.
Exp Hematol ; 40(2): 131-42.e4, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22024107

RESUMO

The pathobiological role of p53 has been widely studied, however, its role in normophysiology is relatively unexplored. We previously showed that p53 knock-down increased ploidy in megakaryocytic cultures. This study aims to examine the effect of p53 loss on in vivo megakaryopoiesis, platelet production, and function, and to investigate the basis for greater ploidy in p53(-/-) megakaryocytic cultures. Here, we used flow cytometry to analyze ploidy, DNA synthesis, and apoptosis in murine cultured and bone marrow megakaryocytes following thrombopoietin administration and to analyze fibrinogen binding to platelets in vitro. Culture of p53(-/-) marrow cells for 6 days with thrombopoietin gave rise to 1.7-fold more megakaryocytes, 26.1% ± 3.6% of which reached ploidy classes ≥64 N compared to 8.2% ± 0.9% of p53(+/+) megakaryocytes. This was due to 30% greater DNA synthesis in p53(-/-) megakaryocytes and 31% greater apoptosis in p53(+/+) megakaryocytes by day 4 of culture. Although the bone marrow and spleen steady-state megakaryocytic content and ploidy were similar in p53(+/+) and p53(-/-) mice, thrombopoietin administration resulted in increased megakaryocytic polyploidization in p53(-/-) mice. Although their platelet counts were normal, p53(-/-) mice exhibited significantly longer bleeding times and p53(-/-) platelets were less sensitive than p53(+/+) platelets to agonist-induced fibrinogen binding and P-selectin secretion. In summary, our in vivo and ex vivo studies indicate that p53 loss leads to increased polyploidization during megakaryopoiesis. Our findings also suggest for the first time a direct link between p53 loss and the development of fully functional platelets resulting in hemostatic deficiencies.


Assuntos
Plaquetas/fisiologia , Trombopoese/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Cruzamento , Tamanho Celular , Hemostasia , Masculino , Camundongos , Selectina-P/metabolismo , Receptores Ativados por Proteinase/agonistas
8.
Blood ; 117(3): 1005-13, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20971951

RESUMO

Two major pathways contribute to Ras-proximate-1-mediated integrin activation in stimulated platelets. Calcium and diacyglycerol-regulated guanine nucleotide exchange factor I (CalDAG-GEFI, RasGRP2) mediates the rapid but reversible activation of integrin αIIbß3, while the adenosine diphosphate receptor P2Y12, the target for antiplatelet drugs like clopidogrel, facilitates delayed but sustained integrin activation. To establish CalDAG-GEFI as a target for antiplatelet therapy, we compared how each pathway contributes to thrombosis and hemostasis in mice. Ex vivo, thrombus formation at arterial or venous shear rates was markedly reduced in CalDAG-GEFI(-/-) blood, even in the presence of exogenous adenosine diphosphate and thromboxane A(2). In vivo, thrombosis was virtually abolished in arterioles and arteries of CalDAG-GEFI(-/-) mice, while small, hemostatically active thrombi formed in venules. Specific deletion of the C1-like domain of CalDAG-GEFI in circulating platelets also led to protection from thrombus formation at arterial flow conditions, while it only marginally increased blood loss in mice. In comparison, thrombi in the micro- and macrovasculature of clopidogrel-treated wild-type mice grew rapidly and frequently embolized but were hemostatically inactive. Together, these data suggest that inhibition of the catalytic or the C1 regulatory domain in CalDAG-GEFI will provide strong protection from athero-thrombotic complications while maintaining a better safety profile than P2Y12 inhibitors like clopidogrel.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Trombose/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Clopidogrel , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fatores de Troca do Nucleotídeo Guanina/sangue , Fatores de Troca do Nucleotídeo Guanina/genética , Hemostasia , Cinética , Masculino , Mesentério/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/irrigação sanguínea , Inibidores da Agregação Plaquetária/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Trombose/tratamento farmacológico , Trombose/genética , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico
9.
J Biol Chem ; 286(5): 3805-14, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21106537

RESUMO

Arrestins can facilitate desensitization or signaling by G protein-coupled receptors (GPCR) in many cells, but their roles in platelets remain uncharacterized. Because of recent reports that arrestins can serve as scaffolds to recruit phosphatidylinositol-3 kinases (PI3K)s to GPCRs, we sought to determine whether arrestins regulate PI3K-dependent Akt signaling in platelets, with consequences for thrombosis. Co-immunoprecipitation experiments demonstrate that arrestin-2 associates with p85 PI3Kα/ß subunits in thrombin-stimulated platelets, but not resting cells. The association is inhibited by inhibitors of P2Y12 and Src family kinases (SFKs). The function of arrestin-2 in platelets is agonist-specific, as PAR4-dependent Akt phosphorylation and fibrinogen binding were reduced in arrestin-2 knock-out platelets compared with WT controls, but ADP-stimulated signaling to Akt and fibrinogen binding were unaffected. ADP receptors regulate arrestin recruitment to PAR4, because co-immunoprecipitates of arrestin-2 with PAR4 are disrupted by inhibitors of P2Y1 or P2Y12. P2Y1 may regulate arrestin-2 recruitment to PAR4 through protein kinase C (PKC) activation, whereas P2Y12 directly interacts with PAR4 and therefore, may help to recruit arrestin-2 to PAR4. Finally, arrestin2(-/-) mice are less sensitive to ferric chloride-induced thrombosis than WT mice, suggesting that arrestin-2 can regulate thrombus formation in vivo. In conclusion, arrestin-2 regulates PAR4-dependent signaling pathways, but not responses to ADP alone, and contributes to thrombus formation in vivo.


Assuntos
Arrestinas/fisiologia , Plaquetas/metabolismo , Receptores Ativados por Proteinase/metabolismo , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais , Animais , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Camundongos , Camundongos Knockout , Transporte Proteico , Trombose/etiologia , beta-Arrestinas
10.
Arterioscler Thromb Vasc Biol ; 29(4): 449-57, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19023091

RESUMO

Platelet G protein-coupled receptors (GPCRs) initiate and reinforce platelet activation and thrombus formation. The clinical utility of antagonists of the P2Y(12) receptor for ADP suggests that other GPCRs and their intracellular signaling pathways may represent viable targets for novel antiplatelet agents. For example, thrombin stimulation of platelets is mediated by 2 protease-activated receptors (PARs), PAR-1 and PAR-4. Signaling downstream of PAR-1 or PAR-4 activates phospholipase C and protein kinase C and causes autoamplification by production of thromboxane A(2), release of ADP, and generation of more thrombin. In addition to ADP receptors, thrombin and thromboxane A(2) receptors and their downstream effectors-including phosphoinositol-3 kinase, Rap1b, talin, and kindlin-are promising targets for new antiplatelet agents. The mechanistic rationale and available clinical data for drugs targeting disruption of these signaling pathways are discussed. The identification and development of new agents directed against specific platelet signaling pathways may offer an advantage in preventing thrombotic events while minimizing bleeding risk.


Assuntos
Plaquetas/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Difosfato de Adenosina/metabolismo , Animais , Plaquetas/enzimologia , Plaquetas/metabolismo , Desenho de Fármacos , Hemorragia/induzido quimicamente , Humanos , Inibidores da Agregação Plaquetária/efeitos adversos , Inibidores da Agregação Plaquetária/química , Antagonistas do Receptor Purinérgico P2 , Receptor PAR-1/antagonistas & inibidores , Receptores Acoplados a Proteínas G/sangue , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12 , Receptores de Trombina/antagonistas & inibidores , Receptores de Tromboxanos/antagonistas & inibidores , Trombina/metabolismo , Tromboxanos/metabolismo
11.
Blood ; 111(7): 3522-30, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18218855

RESUMO

Glycogen synthase kinase (GSK)3beta is a ser-thr kinase that is phosphorylated by the kinase Akt. Although Akt has been shown to regulate platelet function and arterial thrombosis, its effectors in platelets remain unknown. We show here that agonist-dependent phosphorylation of GSK3beta in platelets is Akt dependent. To determine whether GSK3beta regulates platelet function, platelets from mice lacking a single allele of GSK3beta were compared with those of wild-type (WT) controls. GSK3beta+/- platelets demonstrated enhanced agonist-dependent aggregation, dense granule secretion, and fibrinogen binding, compared with WT platelets. Treatment of human platelets with GSK3 inhibitors renders them more sensitive to agonist-induced aggregation, suggesting that GSK3 suppresses platelet function in vitro. Finally, the effect of GSK3beta on platelet function in vivo was evaluated using 2 thrombosis models in mice. In the first, 80% of GSK3beta+/- mice (n=10) formed stable occlusive thrombi after ferric chloride carotid artery injury, whereas the majority of wild-type mice (67%) formed no thrombi (n=15). In a disseminated thrombosis model, deletion of a single allele of GSK3beta in mice conferred enhanced sensitivity to thrombotic insult. Taken together, these results suggest that GSK3beta acts as a negative regulator of platelet function in vitro and in vivo.


Assuntos
Plaquetas/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Agregação Plaquetária , Trombose/etnologia , Animais , Plaquetas/patologia , Lesões das Artérias Carótidas , Cloretos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Compostos Férricos/toxicidade , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Camundongos Knockout , Noxas/toxicidade , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trombose/induzido quimicamente , Trombose/genética , Trombose/patologia
12.
Blood ; 111(7): 3458-67, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18094327

RESUMO

Serglycin (SG), the hematopoietic cell secretory granule proteoglycan, is crucial for storage of specific secretory proteins in mast cells, neutrophils, and cytotoxic T lymphocytes. We addressed the role of SG in platelets using SG-/- mice. Wild-type (WT) but not SG-/- platelets contained chondroitin sulfate proteoglycans. Electron microscopy revealed normal alpha-granule structure in SG-/- platelets. However, SG-/- platelets and megakaryocytes contained unusual scroll-like membranous inclusions, and SG-/- megakaryocytes showed extensive emperipolesis of neutrophils. SG-/- platelets had reduced ability to aggregate in response to low concentrations of collagen or PAR4 thrombin receptor agonist AYPGKF, and reduced fibrinogen binding after AYPGKF, but aggregated normally to ADP. 3H-serotonin and ATP secretion were greatly reduced in SG-/- platelets. The alpha-granule proteins platelet factor 4, beta-thromboglobulin, and platelet-derived growth factor were profoundly reduced in SG-/- platelets. Exposure of P-selectin and alphaIIb after thrombin treatment was similar in WT and SG-/- platelets. SG-/- mice exhibited reduced carotid artery thrombus formation after exposure to FeCl3. This study demonstrates that SG is crucial for platelet function and thrombus formation. We propose that SG-/- platelet function deficiencies are related to inadequate packaging and secretion of selected alpha-granule proteins and reduced secretion of dense granule contents critical for platelet activation.


Assuntos
Plaquetas/metabolismo , Megacariócitos/metabolismo , Agregação Plaquetária , Proteoglicanas/metabolismo , Vesículas Secretórias/metabolismo , Trombose/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Plaquetas/ultraestrutura , Cloretos , Compostos Férricos/farmacologia , Fibrinogênio/genética , Fibrinogênio/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Mastócitos/metabolismo , Mastócitos/patologia , Megacariócitos/ultraestrutura , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/patologia , Noxas/farmacologia , Oligopeptídeos/farmacologia , Selectina-P/genética , Selectina-P/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/genética , Fator Plaquetário 4/genética , Fator Plaquetário 4/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/genética , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Proteoglicanas/genética , Receptores de Trombina/genética , Receptores de Trombina/metabolismo , Vesículas Secretórias/genética , Vesículas Secretórias/ultraestrutura , Serotonina/metabolismo , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia , Trombina/genética , Trombina/metabolismo , Trombose/genética , Trombose/patologia , Proteínas de Transporte Vesicular/genética , beta-Tromboglobulina/genética , beta-Tromboglobulina/metabolismo
13.
J Clin Invest ; 113(3): 441-50, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14755341

RESUMO

Prior studies have shown that PI3Ks play a necessary but incompletely defined role in platelet activation. One potential effector for PI3K is the serine/threonine kinase, Akt, whose contribution to platelet activation was explored here. Two isoforms of Akt were detected in mouse platelets, with expression of Akt2 being greater than Akt1. Deletion of the gene encoding Akt2 impaired platelet aggregation, fibrinogen binding, and granule secretion, especially in response to low concentrations of agonists that activate the G(q)-coupled receptors for thrombin and thromboxane A(2). Loss of Akt2 also impaired arterial thrombus formation and stability in vivo, despite having little effect on platelet responses to collagen and ADP. In contrast, reducing Akt1 expression had no effect except when Akt2 was also deleted. Activation of Akt by thrombin was abolished by deletion of Galpha(q) but was relatively unaffected by deletion of Galpha(i2), which abolished Akt activation by ADP. From these results we conclude that Akt2 is a necessary component of PI3K-dependent signaling downstream of G(q)-coupled receptors, promoting thrombus growth and stability in part by supporting secretion. The contribution of Akt1 is less evident except in the setting in which Akt2 is absent.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/metabolismo , Ativação Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/deficiência , Animais , Tempo de Sangramento , Coagulação Sanguínea/genética , Fibrinogênio/metabolismo , Camundongos , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Isoformas de Proteínas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/metabolismo , Trombose/metabolismo
14.
Blood ; 103(4): 1348-55, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14576067

RESUMO

We have previously shown that platelets express 2 receptor tyrosine kinases, EphA4 and EphB1, and the Eph kinase ligand, ephrinB1, and proposed that transcellular Eph/ephrin interactions made possible by the onset of platelet aggregation promote the further growth and stability of the hemostatic plug. The present study examines how this might occur. The results show that clustering of either ephrinB1 or EphA4 causes platelets to adhere to immobilized fibrinogen via alpha(IIb)beta(3). Adhesion occurs more slowly than with adenosine diphosphate (ADP) and requires phosphatidylinositol 3 (PI3)-kinase and protein kinase C activity but not ephrinB1 phosphorylation. By itself, Eph and ephrin signaling is insufficient to cause aggregation or the binding of soluble fibrinogen, but it can potentiate aggregation initiated by a Ca(++) ionophore or by agonists for thrombin and thromboxane receptors. It also enhances Rap1 activation without requiring ADP secretion, ephrinB1 phosphorylation, or the activation of PI3-kinase and Src. From this we conclude that (1) Eph/ephrin signaling enhances the ability of platelet agonists to cause aggregation provided that those agonists can increase cytosolic Ca(++); (2) this is accomplished in part by activating Rap1; and (3) these effects require oligomerization of ephrinB1 but not phosphotyrosine-based interactions with the ephrinB1 cytoplasmic domain.


Assuntos
Plaquetas/metabolismo , Efrina-B1/metabolismo , Agregação Plaquetária/fisiologia , Transdução de Sinais/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Efrina-A4/genética , Efrina-A4/metabolismo , Humanos , Fosforilação , Adesividade Plaquetária/fisiologia , Proteínas Quinases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
15.
J Biol Chem ; 277(48): 46035-42, 2002 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-12297509

RESUMO

Platelet responses at sites of vascular injury are regulated by intracellular cAMP levels, which rise rapidly when prostacyclin (PGI(2)) is released from endothelial cells. Platelet agonists such as ADP and epinephrine suppress PGI(2)-stimulated cAMP formation by activating receptors coupled to G(i) family members, four of which are present in platelets. To address questions about the specificity of receptor:G protein coupling, the regulation of cAMP formation in vivo and the contribution of G(i)-mediated pathways that do not involve adenylyl cyclase, we studied platelets from mice that lacked the alpha subunits of one or more of the three most abundantly expressed G(i) family members and compared the results with platelets from mice that lacked the PGI(2) receptor, IP. As reported previously, loss of G(i2)alpha or G(z)alpha inhibited aggregation in response to ADP and epinephrine, respectively, producing defects that could not be reversed by adding an adenylyl cyclase inhibitor. Platelets that lacked both G(i2)alpha and G(z)alpha showed impaired responses to both agonists, but the impairment was no greater than in the individual knockouts. Loss of G(i3)alpha had no effect either alone or in combination with G(z)alpha. Loss of either G(z)alpha or G(i2)alpha impaired the ability of ADP and epinephrine to inhibit PGI(2)-stimulated adenylyl cyclase activity and caused a 40%-50% rise in basal cAMP levels, whereas loss of G(i3)alpha did not. Conversely, deletion of IP abolished responses to PGI(2) and caused cAMP levels to fall by 30%, effects that did not translate into enhanced responsiveness to agonists ex vivo. From these results we conclude that 1) cAMP levels in circulating platelets reflect ongoing signaling through G(i2), G(z), and IP, but not G(i3); 2) platelet epinephrine (alpha(2A)-adrenergic) and ADP (P2Y12) receptors display strong preferences among G(i) family members with little evidence of redundancy; and 3) these receptor preferences do not extend to G(i3). Finally, the failure of ADP and epinephrine to inhibit basal, as opposed to PGI(2)-stimulated, cAMP formation highlights the need during platelet activation for G(i) signaling pathways that involve effectors other than adenylyl cyclase.


Assuntos
Adenilil Ciclases/metabolismo , Plaquetas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Transdução de Sinais , Animais , Plaquetas/enzimologia , AMP Cíclico/metabolismo , Epoprostenol/metabolismo , Camundongos
16.
Proc Natl Acad Sci U S A ; 99(14): 9219-24, 2002 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-12084815

RESUMO

Eph kinases are receptor tyrosine kinases whose ligands, the ephrins, are also expressed on the surface of cells. Interactions between Eph kinases and ephrins on adjacent cells play a central role in neuronal patterning and vasculogenesis. Here we examine the expression of ephrins and Eph kinases on human blood platelets and explore their role in the formation of the hemostatic plug. The results show that human platelets express EphA4 and EphB1, and the ligand, ephrinB1. Forced clustering of EphA4 or ephrinB1 led to cytoskeletal reorganization, adhesion to fibrinogen, and alpha-granule secretion. Clustering of ephrinB1 also caused activation of the Ras family member, Rap1B. In platelets that had been activated by ADP and allowed to aggregate, EphA4 formed complexes with two tyrosine kinases, Fyn and Lyn, and the cell adhesion molecule, L1. Blockade of Eph/ephrin interactions prevented the formation of these complexes and caused platelet aggregation at low ADP concentrations to become more readily reversible. We propose that when sustained contacts between platelets have occurred in response to agonists such as collagen, ADP, and thrombin, the binding of ephrins to Eph kinases on adjacent platelets provides a mechanism to perpetuate signaling and promote stable platelet aggregation.


Assuntos
Proteínas de Membrana/sangue , Agregação Plaquetária/fisiologia , Receptores Proteína Tirosina Quinases/sangue , Difosfato de Adenosina/farmacologia , Plaquetas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Colágeno/farmacologia , Efrina-B1 , Proteínas Fetais , Fibrinogênio/metabolismo , Hemostasia , Humanos , Técnicas In Vitro , Complexo Antígeno L1 Leucocitário , Glicoproteínas de Membrana/sangue , Modelos Biológicos , Moléculas de Adesão de Célula Nervosa/sangue , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas/sangue , Proteínas Proto-Oncogênicas c-fyn , Receptor EphA4 , Receptores da Família Eph , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Trombina/farmacologia , Proteínas rap de Ligação ao GTP/sangue , Quinases da Família src/sangue
17.
J Biol Chem ; 277(26): 23382-90, 2002 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-11970953

RESUMO

It has become increasingly appreciated that receptors coupled to G(alpha)(i) family members can stimulate platelet aggregation, but the mechanism for this has remained unclear. One possible mediator is the small GTPase, Rap1, which has been shown to contribute to integrin activation in several cell lines and to be activated by a calcium-dependent mechanism in platelets. Here, we demonstrate that Rap1 is also activated by G(alpha)(i) family members in platelets. First, we show that platelets from mice lacking the G(alpha)(i) family member G(alpha)(z) (which couples to the alpha(2A) adrenergic receptor) are deficient in epinephrine-stimulated Rap1 activation. We also show that platelets from mice lacking G(alpha)(i2), which couples to the ADP receptor, P2Y12, exhibit reduced Rap1 activation in response to ADP. In contrast, platelets from mice that lack G(alpha)(q) show no decrease in the ability to activate Rap1 in response to epinephrine but show a partial reduction in ADP-stimulated Rap1 activation. This result, combined with studies of human platelets treated with ADP receptor-selective inhibitors, indicates that ADP-stimulated Rap1 activation in human platelets is dependent on both the G(alpha)(i)-coupled P2Y12 receptor and the G(alpha)(q)-coupled P2Y1 receptor. G(alpha)(i)-dependent activation of Rap1 in platelets does not appear to be mediated by enhanced intracellular calcium release because no increase in intracellular calcium concentration was detected in response to epinephrine and because the calcium response to ADP was not diminished in platelets from the G(alpha)(i2)-/- mouse. Finally, using human platelets treated with selective inhibitors of phosphatidylinositol 3-kinase (PI3K) and mouse platelets selectively lacking the G(beta)(gamma)-activated form of his enzyme (PI3Kgamma), we show that G(i)-mediated Rap1 activation is PI3K-dependent. In summary, activation of Rap1 can be stimulated by G(alpha)(i)- and PI3K-dependent mechanisms in platelets and by G(q)- and Ca(2+)-dependent mechanisms, both of which may play a role in promoting platelet activation.


Assuntos
Plaquetas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Membrana , Proteínas rap1 de Ligação ao GTP/metabolismo , Difosfato de Adenosina/farmacologia , Adenilil Ciclases/fisiologia , Animais , Epinefrina/farmacologia , Guanosina Trifosfato/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Receptores Purinérgicos P2/fisiologia , Receptores Purinérgicos P2Y12
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA