Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Signal ; 50: 111-120, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29793021

RESUMO

Phosphoinositide 3-kinase (PI3K) plays an important role in platelet function and contributes to platelet hyperreactivity induced by elevated levels of circulating peptide hormones, including thrombopoietin (TPO). Previous work established an important role for the PI3K isoform; p110ß in platelet function, however the role of p110α is still largely unexplored. Here we sought to investigate the role of p110α in TPO-mediated hyperactivity by using a conditional p110α knockout (KO) murine model in conjunction with platelet functional assays. We found that TPO-mediated enhancement of collagen-related peptide (CRP-XL)-induced platelet aggregation and adenosine triphosphate (ATP) secretion were significantly increased in p110α KO platelets. Furthermore, TPO-mediated enhancement of thrombus formation by p110α KO platelets was elevated over wild-type (WT) platelets, suggesting that p110α negatively regulates TPO-mediated priming of platelet function. The enhancements were not due to increased flow through the PI3K pathway as phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P3) formation and phosphorylation of Akt and glycogen synthase kinase 3 (GSK3) were comparable between WT and p110α KO platelets. In contrast, extracellular responsive kinase (ERK) phosphorylation and thromboxane (TxA2) formation were significantly enhanced in p110α KO platelets, both of which were blocked by the MEK inhibitor PD184352, whereas the p38 MAPK inhibitor VX-702 and p110α inhibitor PIK-75 had no effect. Acetylsalicylic acid (ASA) blocked the enhancement of thrombus formation by TPO in both WT and p110α KO mice. Together, these results demonstrate that p110α negatively regulates TPO-mediated enhancement of platelet function by restricting ERK phosphorylation and TxA2 synthesis in a manner independent of its kinase activity.


Assuntos
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Ativação Plaquetária/fisiologia , Trombopoetina/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trombose/metabolismo , Tromboxano A2/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Platelets ; 27(4): 286-94, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26587753

RESUMO

Platelets are critical for maintaining vascular hemostasis, but also play a major role in the formation of occlusive cardiovascular and cerebrovascular thrombi under disease conditions. Secretion of platelet alpha and dense granules is a requirement for efficient thrombus formation. Understanding and targeting the mechanisms of secretion is important to aid the development of effective antithrombotics. SNAP29 is a tSNARE found in platelets, but whose role has not been defined. Using a platelet-specific SNAP29 knockout mouse model, we assessed the role of SNAP29 in platelet secretion and function under standardized conditions and also in in vitro and in vivo thrombosis. The data showed no major defects in SNAP29-null platelets, but revealed a minor defect in α-granule secretion and a significant increase in embolization rate of thrombi in vivo. These data suggest that SNAP29 contributes to the regulation of platelet α-granule secretion and thrombus stability, possibly partially masked by functional redundancy with other tSNAREs, such as SNAP23.


Assuntos
Plaquetas/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteínas SNARE/metabolismo , Animais , Biomarcadores , Plaquetas/ultraestrutura , Ordem dos Genes , Loci Gênicos , Camundongos , Camundongos Knockout , Adesividade Plaquetária , Agregação Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ligação Proteica , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Transdução de Sinais , Trombose/genética , Trombose/metabolismo , Trombose/patologia
3.
J Thromb Haemost ; 13(11): 2102-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26334261

RESUMO

BACKGROUND: RhoA is an important regulator of platelet responses downstream of Gα13 , yet we still know little about its regulation in platelets. Leukemia-associated Rho guanine-nucleotide exchange factor (GEF [LARG]), a RhoA GEF, is highly expressed in platelets and may constitute a major upstream activator of RhoA. To this end, it is important to determine the role of LARG in platelet function and thrombosis. METHODS AND RESULTS: Using a platelet-specific gene knockout, we show that the absence of LARG results in a marked reduction in aggregation and dense-granule secretion in response to the thromboxane mimetic U46619 and proteinase-activated receptor 4-activating peptide, AYPGKF, but not to adenosine diphosphate. In a ferric chloride thrombosis model in vivo, this translated into a defect, under mild injury conditions. Importantly, agonist-induced RhoA activation was not affected by the absence of LARG, although basal activity was reduced, suggesting that LARG may play a housekeeper role in regulating constitutive RhoA activity. CONCLUSIONS: LARG plays an important role in platelet function and thrombosis in vivo. However, although LARG may have a role in regulating the resting activation state of RhoA, its role in regulating platelet function may principally be through RhoA-independent pathways, possibly through other Rho family members.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária/fisiologia , Fatores de Troca de Nucleotídeo Guanina Rho/fisiologia , Trombose/sangue , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Cloretos/toxicidade , Cruzamentos Genéticos , Compostos Férricos/toxicidade , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Oligopeptídeos/farmacologia , Especificidade de Órgãos , Agregação Plaquetária , Fatores de Troca de Nucleotídeo Guanina Rho/sangue , Fatores de Troca de Nucleotídeo Guanina Rho/deficiência , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Trombose/induzido quimicamente
4.
J Thromb Haemost ; 12(5): 726-35, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24581425

RESUMO

BACKGROUND: Protein kinase C (PKC) is a major regulator of platelet function and secretion. The underlying molecular pathway from PKC to secretion, however, is poorly understood. By a proteomics screen we identified the guanine nucleotide exchange factor cytohesin-2 as a candidate PKC substrate. OBJECTIVES: We aimed to validate cytohesin-2 as a PKC substrate in platelets and to determine its role in granule secretion and other platelet responses. METHODS AND RESULTS: Immunoprecipitation was performed with a phosphoserine PKC substrate antibody followed by mass spectrometry, leading to the identification of cytohesin-2. By western blotting we showed that different agonists induced cytohesin-2 phosphorylation by PKC. Protein function was investigated using a pharmacological approach. The cytohesin inhibitor SecinH3 significantly enhanced platelet dense granule secretion and aggregation, as measured by lumi-aggregometry. Flow cytometry data indicate that α-granule release and integrin αII b ß3 activation were not affected by cytohesin-2 inhibition. Lysosome secretion was assessed by a colorimetric assay and was also unchanged. As shown by western blotting, ARF6 interacted with cytohesin-2 and was present in an active GTP-bound form under basal conditions. Upon platelet stimulation, this interaction was largely lost and ARF6 activation decreased, both of which could be rescued by PKC inhibition. CONCLUSIONS: Cytohesin-2 constitutively suppresses platelet dense granule secretion and aggregation by keeping ARF6 in a GTP-bound state. PKC-mediated phosphorylation of cytohesin-2 relieves this inhibitory effect, thereby promoting platelet secretion and aggregation.


Assuntos
Actinas/metabolismo , Plaquetas/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Proteína Quinase C/metabolismo , Fator 6 de Ribosilação do ADP , Trifosfato de Adenosina/metabolismo , Animais , Plaquetas/citologia , Citometria de Fluxo , Guanosina Trifosfato/metabolismo , Humanos , Imunoprecipitação , Lisossomos/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Knockout , Fosforilação , Ativação Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteômica , Vesículas Secretórias/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
5.
Cell Death Dis ; 4: e969, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24357800

RESUMO

Platelets enhance thrombin generation at sites of vascular injury by exposing phosphatidylserine during necrosis-like cell death. Anoctamin 6 (Ano6) is required for Ca(2+)-dependent phosphatidylserine exposure and is defective in patients with Scott syndrome, a rare bleeding disorder. Ano6 may also form Cl(-) channels, though the role of Cl(-) fluxes in platelet procoagulant activity has not been explored. We found that Cl(-) channel blockers or removal of extracellular Cl(-) inhibited agonist-induced phosphatidylserine exposure. However, this was not due to direct inhibition of Ca(2+)-dependent scrambling since Ca(2+) ionophore-induced phosphatidylserine exposure was normal. This implies that the role of Ano6 in Ca(2+-)dependent PS exposure is likely to differ from any putative function of Ano6 as a Cl(-) channel. Instead, Cl(-) channel blockade inhibited agonist-induced Ca(2+) entry. Importantly, Cl(-) channel blockers also prevented agonist-induced membrane hyperpolarization, resulting in depolarization. We propose that Cl(-) entry through Cl(-) channels is required for this hyperpolarization, maintaining the driving force for Ca(2+) entry and triggering full phosphatidylserine exposure. This demonstrates a novel role for Cl(-) channels in controlling platelet death and procoagulant activity.


Assuntos
Plaquetas/metabolismo , Cálcio/metabolismo , Canais de Cloreto/metabolismo , Fosfatidilserinas/metabolismo , Trombose/metabolismo , Anoctaminas , Plaquetas/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Células Cultivadas , Citometria de Fluxo , Humanos , Necrose/induzido quimicamente , Necrose/metabolismo , Peptídeos/farmacologia , Proteínas de Transferência de Fosfolipídeos , Trombina/farmacologia
7.
J Thromb Haemost ; 9(12): 2457-65, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21951302

RESUMO

BACKGROUND: The zebrafish (Danio rerio) is becoming an attractive model organism for the assessment of gene function in thrombosis in vivo. Zebrafish, as a thrombosis model, have several advantages, with the capacity to follow thrombus formation at high resolution in real time using intravital microscopy, without the need for complex surgical techniques, and the capability to rapidly knockdown gene expression using morpholino antisense approaches. OBJECTIVES: We have recently shown, in mouse models, that protein kinase C alpha (PKCα) plays a critical role in regulating thrombus formation in vivo. PKC beta (ß) plays a non-redundant role also in platelet function in vitro, but the function of this gene had not yet been assessed in vivo. METHODS: In the present study, we analyzed the function of both PKCα and PKCß in the zebrafish model in vivo, by live imaging using a laser-induced injury of the main caudal artery in 3-day-old larvae. RESULTS: We showed that D. rerio express orthologs of both the PKCα and PKCß genes, with high sequence identity. Translation blocking and splice-blocking morpholinos effectively and specifically knockdown expression of these genes and knockdown with either morpholino leads to attenuated thrombus formation, as assessed by several quantitative parameters including time to initial adhesion and peak thrombus surface area. CONCLUSIONS: Our data indicate that these two highly related genes play non-redundant roles in regulating thrombosis, an observation that supports our previous in vitro murine data, and suggests unique roles, and possibly unique regulation, for PKCα and PKCß in controlling platelet function in vivo.


Assuntos
Modelos Animais de Doenças , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C/metabolismo , Trombose/enzimologia , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Técnicas de Silenciamento de Genes , Proteína Quinase C/genética , Proteína Quinase C beta , Proteína Quinase C-alfa/genética
8.
J Thromb Haemost ; 9(8): 1599-607, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21649850

RESUMO

BACKGROUND: Cytosolic calcium concentration is a critical regulator of platelet activation, and so platelet Ca(2+) signaling must be tightly controlled. Thrombin-induced Ca(2+) signaling is enhanced by inhibitors of protein kinase C (PKC), suggesting that PKC negatively regulates the Ca(2+) signal, although the mechanisms by which this occurs and its physiological relevance are still unclear. OBJECTIVES: To investigate the mechanisms by which PKC inhibitors enhance thrombin-induced Ca(2+) signaling, and to determine the importance of this pathway in platelet activation. METHODS: Cytosolic Ca(2+) signaling was monitored in fura-2-loaded human platelets. Phosphatidylserine (PS) exposure, a marker of platelet procoagulant activity, was measured by annexin V binding and flow cytometry. RESULTS: PKC inhibition by bisindolylmaleimide-I (BIM-I) enhanced α-thrombin-induced Ca(2+) signaling in a concentration-dependent manner. PAR1 signaling, activated by SFLLRN, was enhanced much more strongly than PAR4, activated by AYPGKF or γ-thrombin, which is a potent PAR4 agonist but a poor activator of PAR1. BIM-I had little effect on α-thrombin-induced signaling following treatment with the PAR1 antagonist, SCH-79797. BIM-I enhanced Ca(2+) release from intracellular stores and Ca(2+) entry, as assessed by Mn(2+) quench. However, the plasma membrane Ca(2+) ATPase inhibitor, 5(6)-carboxyeosin, did not prevent the effect of BIM-I. PKC inhibition strongly enhanced α-thrombin-induced PS exposure, which was reversed by blockade of PAR1. CONCLUSIONS: Together, these data show that when PAR1 is stimulated, PKC negatively regulates Ca(2+) release and Ca(2+) entry, which leads to reduced platelet PS exposure.


Assuntos
Plaquetas/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Indóis/farmacologia , Maleimidas/farmacologia , Fosfatidilserinas/metabolismo , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Receptor PAR-1/efeitos dos fármacos , Receptores de Trombina/efeitos dos fármacos , Anexina A5/metabolismo , Plaquetas/enzimologia , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Proteína Quinase C/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Trombina/metabolismo , Fatores de Tempo
9.
J Thromb Haemost ; 8(3): 567-76, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20002543

RESUMO

BACKGROUND: The integrin alpha(IIb)beta(3) is the major mediator of platelet aggregation and has, therefore, become an important target of antithrombotic therapy. Antagonists of alpha(IIb)beta(3), for example abciximab, tirofiban and eptifibatide, are used in the treatment of acute coronary syndromes. However, in addition to effective blockade of the integrin, binding of can induce conformational changes in the integrin and can also induce integrin clustering. This class effect of RGD-ligand mimetics might, therefore, underlie paradoxical platelet activation and thrombosis previously reported. OBJECTIVES: To examine the components of signaling pathways and functional responses in platelets that may underlie this phenomenon of paradoxical platelet activation. METHODS: We assessed the effect of lotrafiban, and other alpha(IIb)beta(3) antagonists including the clinically used drug tirofiban, on tyrosine phosphorylation of key signaling proteins in platelets by immunoblotting and also platelet functional outputs such as cytosolic calcium responses, phosphatidylserine exposure (pro-coagulant activity) and dense granule release. RESULTS: In all cases, no effect of alpha(IIb)beta(3) antagonists were observed on their own, but these integrin antagonists did lead to a marked potentiation of glycoprotein VI (GPVI)-associated FcR gamma-chain phosphorylation, activation of Src family kinases and Syk kinase. This correlated with increased dense granule secretion, cytosolic calcium response and exposure of phosphatidylserine on the platelet surface. P2Y(12) antagonism abolished the potentiated phosphatidylserine exposure and dense granule secretion but not the cytosolic calcium response. CONCLUSIONS: These data provide a mechanism for enhancement of platelet activity by alpha(IIb)beta(3) inhibitors, but also reveal a potentially important signaling pathway operating from the integrin to GPVI signaling.


Assuntos
Biomimética , Plaquetas/efeitos dos fármacos , Oligopeptídeos/sangue , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/metabolismo , Benzodiazepinas/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Ativação Enzimática , Humanos , Cadeias gama de Imunoglobulina/sangue , Peptídeos e Proteínas de Sinalização Intracelular/sangue , Ligantes , Fosfatidilserinas/sangue , Fosforilação , Piperidinas/farmacologia , Inibidores da Agregação Plaquetária/efeitos adversos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Conformação Proteica , Proteínas Tirosina Quinases/sangue , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/sangue , Receptores Purinérgicos P2Y12 , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Quinase Syk , Fatores de Tempo , Tirofibana , Tirosina/análogos & derivados , Tirosina/sangue , Tirosina/farmacologia , Quinases da Família src/sangue
10.
J Thromb Haemost ; 8(3): 454-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20002545

RESUMO

Platelet activation is a complex balance of positive and negative signaling pathways. The protein kinase C (PKC) family is a major regulator of platelet granule secretion, integrin activation, aggregation, spreading and procoagulant activity. As broad-spectrum PKC inhibitors reduce secretion and aggregation, the PKC family is generally considered to be a positive regulator of platelet activation. However, the individual members of the PKC family that are expressed in platelets are regulated in different ways, and an increasing body of evidence indicates that they have distinct, and often opposing, roles. Many of the recent advances in understanding the contributions of individual PKC isoforms have come from mouse gene knockout studies. PKCalpha, a classic isoform, is an essential positive regulator of granule secretion and thrombus formation, both in vitro and in vivo. Mice lacking PKCalpha show much reduced thrombus formation in vivo but do not have a bleeding defect, suggesting that PKCalpha could be an attractive antithrombotic target. Important, apparently non-redundant, roles, both positive and negative, for the novel PKC isoforms delta, theta and epsilon in granule secretion have also been proposed, indicating highly complex regulation of this essential process. Similarly, PKCbeta, PKCdelta and PKCtheta have non-redundant roles in platelet spreading, as absence of either PKCbeta or PKCtheta reduces spreading, whereas PKCdelta negatively regulates filopodial formation. This negative signaling by PKCdelta may reduce platelet aggregation and so restrict thrombus formation. In this review, we discuss the current understanding of the regulation and functions of individual PKC isoforms in platelet activation and thrombus formation.


Assuntos
Plaquetas/enzimologia , Ativação Plaquetária , Proteína Quinase C/metabolismo , Trombose/enzimologia , Animais , Grânulos Citoplasmáticos/enzimologia , Ativação Enzimática , Humanos , Isoenzimas , Camundongos , Camundongos Knockout , Ativação Plaquetária/genética , Agregação Plaquetária , Proteína Quinase C/genética , Transdução de Sinais , Trombose/sangue , Trombose/genética , Tromboxanos/metabolismo
11.
J Thromb Haemost ; 6(8): 1393-404, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18513210

RESUMO

BACKGROUND: Adenosine diphosphate (ADP) is a critical regulator of platelet activation, mediating its actions through two G protein-coupled receptors (GPCRs), the P2Y(1) and P2Y(12) purinergic receptors. Recently, we demonstrated that both receptors desensitize and internalize in human platelets by differential kinase-dependent mechanisms. OBJECTIVES: To demonstrate whether responses to P2Y(1) and P2Y(12) purinergic receptors resensitize in human platelets and determine the role of receptor traffic in this process. METHODS: These studies were undertaken either in human platelets or in cells stably expressing epitope-tagged P2Y(1) and P2Y(12) purinergic receptor constructs. RESULTS: In this study we show for the first time that responses to both of these receptors can rapidly resensitize following agonist-dependent desensitization in human platelets. Further, we show that in human platelets or in 1321N1 cells stably expressing receptor constructs, the disruption of receptor internalization, dephosphorylation or subsequent receptor recycling is sufficient to block resensitization of purinergic receptor responses. We also show that, in platelets, internalization of both these receptors is dependent upon dynamin, and that this process is required for resensitization of responses. CONCLUSIONS: This study is therefore the first to show that both P2Y(1) and P2Y(12) receptor activities are rapidly and reversibly modulated in human platelets, and it reveals that the underlying mechanism requires receptor trafficking as an essential part of this process.


Assuntos
Plaquetas/metabolismo , Receptores Purinérgicos P2/sangue , Receptores Purinérgicos/sangue , Difosfato de Adenosina/sangue , Difosfato de Adenosina/farmacologia , Plaquetas/efeitos dos fármacos , Cálcio/sangue , Linhagem Celular , AMP Cíclico/sangue , Humanos , Técnicas In Vitro , Monensin/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Agonistas Purinérgicos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
12.
J Thromb Haemost ; 6(3): 534-43, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18088343

RESUMO

BACKGROUND: Thromboxane A(2) and ADP are two major platelet agonists that stimulate two sets of G protein-coupled receptors to activate platelets. Although aggregation responses to ADP and thromboxane desensitize, there are no reports currently addressing whether activation by one agonist may heterologously desensitize responses to the other. OBJECTIVES: To demonstrate whether responses to ADP or U46619 may be modulated by prior treatment of platelets with the alternate agonist, revealing a level of cross-desensitization between receptor systems. RESULTS: Here we show that pretreatment of platelets with either agonist substantially desensitizes aggregation responses to the other agonist. Calcium responses to thromboxane receptor activation are desensitized by preactivation of P2Y(1) but not P2Y(12) receptors. This heterologous desensitization is mediated by a protein kinase C (PKC)-independent mechanism. Reciprocally, calcium responses to ADP are desensitized by pretreatment of platelets with the thromboxane analogue, U46619, and P2Y(12)-mediated inhibition of adenylate cyclase is also desensitized by pretreatment with U46619. In this direction, desensitization is comprised of two components, a true heterologous component that is PKC-independent, and a homologous component that is mediated through stimulated release of dense granule ADP. CONCLUSIONS: This study reveals cross-desensitization between ADP and thromboxane receptor signaling in human platelets. Cross-desensitization is mediated by protein kinases, involving PKC-dependent and independent pathways, and indicates that alterations in the activation state of one receptor may have effects upon the sensitivity of the other receptor system.


Assuntos
Plaquetas/metabolismo , Agregação Plaquetária , Receptores Purinérgicos P2/química , Receptores de Tromboxanos/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/química , Cálcio/metabolismo , AMP Cíclico/metabolismo , Citosol/metabolismo , Humanos , Ativação Plaquetária , Proteína Quinase C/metabolismo , Serotonina/química , Transdução de Sinais , Tromboxanos/química
13.
Biochem Soc Trans ; 35(Pt 5): 1005-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956264

RESUMO

Platelets are central to haemostasis and thrombosis. Many key steps in platelet activation and aggregation are regulated by members of the PKC (protein kinase C) family. Multiple isoforms of PKC are expressed in platelets, and evidence is emerging that different isoforms play distinct roles in the platelet activation process. This may, in part, be regulated by isoform-specific interactions between PKC family members and other intracellular signalling molecules, such as tyrosine kinases, or the actin cytoskeleton regulator, VASP (vasodilator-stimulated phosphoprotein). The contributions of individual PKC isoforms can be addressed directly in platelets from knockout mouse models, which are providing key insights into the physiological function of PKC isoform diversity and can be a valuable complimentary approach to more commonly used pharmacological analyses. Using knockout mouse models, recent reports have demonstrated the importance of PKCbeta and PKCtheta in integrin-dependent platelet spreading, and also a novel role for PKCdelta in regulating filopodial formation, highlighting the utility of such models to investigate the functions of specific PKC isoforms in a physiological process that is significant to our understanding of cardiovascular disease.


Assuntos
Plaquetas/enzimologia , Isoenzimas/sangue , Proteína Quinase C/sangue , Animais , Humanos , Camundongos , Modelos Animais , Ativação Plaquetária
14.
Blood ; 97(12): 3989-91, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11389045

RESUMO

The glycoprotein (GP)-Ib-IX-V receptor complex has recently been reported to signal through a pathway similar to that used by the collagen receptor GPVI, with a critical role described for the Fc receptor gamma-chain. The evidence for this was based in part on studies with the GPIbalpha-selective snake venom toxin, alboaggregin-A. In the present study, it is reported that alboaggregin-A has activity at the collagen receptor GPVI in addition to GPIbalpha, and evidence is provided that this contributes to protein tyrosine phosphorylation, shape change, and GPIIb-IIIa-dependent aggregation. This may explain why responses to alboaggregin-A are distinct from those to von Willebrand factor-ristocetin. (Blood. 2001;97:3989-3991)


Assuntos
Venenos de Crotalídeos/farmacologia , Glicoproteínas da Membrana de Plaquetas/efeitos dos fármacos , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Venenos de Crotalídeos/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/farmacologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/fisiologia , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo
15.
FEBS Lett ; 459(1): 27-32, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10508911

RESUMO

SH2 domain containing phosphatase-2 (SHP-2) has an important regulatory role in a variety of cell types. However, little is known concerning its function in platelets. We show here that, in thrombin-stimulated human platelets, SHP-2 undergoes a time-dependent association with platelet endothelial cell adhesion molecule-1 (PECAM-1) and four low molecular weight phosphoproteins which are attenuated by the Src kinase inhibitor PP1. The low molecular weight proteins, which may be transmembrane proteins, are shown to bind exclusively to the N-terminal SH2 domain of SHP-2 and are therefore possible activators of the phosphatase. In addition, SHP-2 phosphatase activity is shown to be increased following thrombin stimulation or cross-linking of PECAM.


Assuntos
Plaquetas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Trombina/metabolismo , Sítios de Ligação , Reagentes de Ligações Cruzadas , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Peso Molecular , Fosforilação , Ativação Plaquetária , Proteína Fosfatase 2 , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Especificidade por Substrato , Tirosina/metabolismo , Domínios de Homologia de src , Quinases da Família src/metabolismo
16.
Blood ; 94(5): 1648-56, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10477689

RESUMO

The adhesion molecule von Willebrand factor (vWF) activates platelets upon binding 2 surface receptors, glycoprotein (GP) Ib-V-IX and integrin alpha(IIb)beta(3). We have used 2 approaches to selectively activate GP Ib using either the snake venom lectin alboaggregin-A or mutant recombinant forms of vWF (triangle upA1-vWF and RGGS-vWF) with selective binding properties to its 2 receptors. We show that activation of GP Ib induces platelet aggregation, secretion of 5-hydroxy tryptamine (5-HT), and an increase in cytosolic calcium. Syk becomes tyrosine phosphorylated and activated downstream of GP Ib, and associates with several tyrosine-phosphorylated proteins including the Fc receptor gamma-chain through interaction with Syk SH2 domains. GP Ib physically associates with the gamma-chain in GST-Syk-SH2 precipitates from platelets stimulated through GP Ib, and 2 Src family kinases, Lyn and Fyn, also associate with this signaling complex. In addition, GP Ib stimulation couples to tyrosine phosphorylation of phospholipase Cgamma2. The Src family-specific inhibitor PP1 dose-dependently inhibits phosphorylation of Syk, its association with tyrosine-phosphorylated gamma-chain, phosphorylation of PLCgamma2, platelet aggregation, and 5-HT release. The results indicate that, upon activation, GP Ib is physically associated with FcR gamma-chain and members of the Src family kinases, leading to phosphorylation of the gamma-chain, recruitment, and activation of Syk. Phosphorylation of PLCgamma2 also lies downstream of Src kinase activation and may critically couple early signaling events to functional platelet responses.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Fc/metabolismo , Quinases da Família src/metabolismo , Humanos , Complexo Glicoproteico GPIb-IX de Plaquetas/química , Proteínas Proto-Oncogênicas c-fyn , Agregação de Receptores , Transdução de Sinais , Fator de von Willebrand/metabolismo
17.
Br J Pharmacol ; 115(1): 101-6, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7647963

RESUMO

1. There is controversy in the literature as to whether collagen is able to induce directly a rise in cytosolic calcium concentration ([Ca2+]i) in human platelets. We have addressed this question by observing the cytosolic calcium response of single fura-2-loaded human platelets settling onto a collagen-coated surface using dynamic fluorescence ratio imaging. 2. Following a short lag phase after adherence to collagen fibres, platelets underwent a rapid rise in cytosolic calcium from basal values of 80 +/- 13 nM (n = 24) to a peak of 475 +/- 42 nM (n = 24) which was sustained for the remaining period of the experiment. 3. The tyrphostin protein tyrosine kinase inhibitor, ST271, reduced substantially the proportion of platelets which exhibited a rise in [Ca2+]i on adherence to collagen and transformed the response in remaining cells to one of oscillations. 4. In contrast, and as a control for collagen, laminin-coated surfaces induced adherence of human platelets without elevating intracellular [Ca2+]; the cells however remained responsive to ADP. 5. We conclude that collagen directly induces a rise in cytosolic calcium in single human platelets through a tyrosine kinase-mediated pathway.


Assuntos
Plaquetas/metabolismo , Cálcio/metabolismo , Colágeno/farmacologia , Difosfato de Adenosina/farmacologia , Plaquetas/efeitos dos fármacos , Adesão Celular/fisiologia , Citosol/metabolismo , Humanos , Técnicas In Vitro , Fosfatos de Inositol/biossíntese , Laminina/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Estirenos/farmacologia
18.
J Vet Pharmacol Ther ; 17(5): 374-8, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7853463

RESUMO

The responses of equine blood platelets in citrated platelet-rich plasma to arachidonic acid, U44069 (prostaglandin endoperoxide analogue), adenosine 5'-diphosphate, platelet-activating factor or collagen were investigated by turbidimetric aggregometry. Pre-treatment of the platelets with aspirin (1 mmol/l) or paracetamol (1.3 mmol/l) abolished shape change and aggregation in response to arachidonic acid; decreased the rate of aggregation in response to collagen, with no separate effect on shape change; had no marked effect on aggregation caused by the other agonists; but in no case transformed irreversible aggregation to reversible aggregation. We conclude that thromboxane A2 generation is of minor importance in the aggregation of equine platelets, and in particular that thromboxane A2 is not a significant mediator of irreversible aggregation.


Assuntos
Acetaminofen/farmacologia , Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Cavalos/sangue , Agregação Plaquetária/efeitos dos fármacos , Acetaminofen/administração & dosagem , Difosfato de Adenosina/farmacologia , Animais , Ácido Araquidônico/farmacologia , Aspirina/administração & dosagem , Tamanho Celular/efeitos dos fármacos , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Técnicas In Vitro , Nefelometria e Turbidimetria/veterinária , Fator de Ativação de Plaquetas/farmacologia , Endoperóxidos Sintéticos de Prostaglandinas/farmacologia , Tromboxano A2/metabolismo
19.
Res Vet Sci ; 54(2): 235-43, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8460266

RESUMO

Pre-incubation of equine platelets in platelet-rich plasma with adenosine 5'-diphosphate (ADP) induced a reduction in aggregation responsiveness to subsequent additions of ADP. The desensitisation was shown to be homologous since the responsiveness to platelet-activating factor, thrombin, collagen, 5-hydroxytryptamine or ionomycin remained unchanged. Adenosine 5'-(beta-thio)-diphosphate (ADP beta S), a non-hydrolysable analogue of ADP, was shown to act as an agonist inducing aggregation by interaction with the ADP receptor. ADP beta S was then used in the desensitisation studies in which residual ADP was degraded by the addition of apyrase. The desensitisation to ADP beta S fully recovered by one hour after pre-treatment with ADP and was not induced by an extracellular mediator. The mechanism of desensitisation is therefore likely to involve the ADP receptor or proximal intermediates in the signal transduction pathway for ADP.


Assuntos
Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Cavalos/sangue , Agregação Plaquetária/efeitos dos fármacos , Tionucleotídeos/farmacologia , Monofosfato de Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Plaquetas/fisiologia , Receptores Purinérgicos/metabolismo
20.
Equine Vet J ; 25(1): 45-8, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8422884

RESUMO

Equine platelets in platelet-rich plasma were incubated with the fluorescent indicator dye, Fura-2-AM (Fura-2-acetoxymethyl ester) and the degree of loading of the cells with the dye and the extent of hydrolysis of the ester was assessed by quantitative fluorimetry and by thin-layer chromatography respectively. Under these conditions the cells loaded poorly with Fura-2 to a concentration of 4 microM. The technique was validated by demonstrating adequate loading of human platelets with Fura-2, to a concentration of 250-300 microM, using the same method. The removal of plasma from the extracellular medium was important for successful loading of the cells with the dye since washed cells resuspended in a physiological salt solution loaded adequately with Fura-2 to a concentration of 190 microM. Cells loaded as such showed a resting [Ca2+]i of 129 nM and a concentration-dependent rise in [Ca2+]i to a transient maximum of 350 nM when stimulated by the pro-aggregatory agonist adenosine 5'-diphosphate (ADP).


Assuntos
Plaquetas/metabolismo , Cálcio/sangue , Corantes Fluorescentes , Fura-2/análogos & derivados , Cavalos/sangue , Difosfato de Adenosina/metabolismo , Animais , Cromatografia em Camada Fina/veterinária , Fluorometria/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA