Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Thromb Haemost ; 105(3): 479-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21136013

ABSTRACT

We have recently shown that ADP-induced activation of protein kinase C (PKC) requires the co-stimulation of both P2Y1 and P2Y12 receptors. In this work, we show that inhibition of ADP-mediated phosphorylation of pleckstrin, the main PKC substrate, caused by antagonists of the P2Y12 receptor can be reversed by stimulation of the α2-adrenergic receptor by epinephrine. However, we also observed that addition of epinephrine alone caused a marked phosphorylation of pleckstrin. This effect occurred in the absence of Gq stimulation, as it was not associated to intracellular Ca2+ release. Epinephrine-induced pleckstrin phosphorylation was time- and dose-dependent, and was inhibited by the α2-adrenergic antagonist yohimbin. Phosphorylation of pleckstrin did not occur when platelet stimulation with epinephrine was performed in the presence of the ADP scavenger apyrase, and was suppressed by antagonists of both P2Y1 and P2Y12 ADP receptors. Importantly, no release of dense granules was measured in epinephrine-treated platelets. Addition of epinephrine to platelets was also able to stimulate Rap1b activation. Similarly to pleckstrin phosphorylation, however, this effect was prevented in the presence of apyrase or upon pharmacologic blockade of either P2Y1 or P2Y12 receptors. These results indicate that sub-threshold amounts of ADP in the medium are essential to allow epinephrine stimulation of α2-adrenergic receptor to elicit platelet responses, and reveal a novel synergism among strong stimulation of Gz and sub-threshold stimulation of both Gq and Gi, able to dissociate PKC activation from intracellular Ca2+ mobilisation.


Subject(s)
Epinephrine/chemistry , Receptors, Purinergic P2Y12/metabolism , Receptors, Purinergic P2Y1/metabolism , Blood Proteins/chemistry , Calcium/chemistry , Cytosol/metabolism , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Humans , Phosphoproteins/chemistry , Phosphorylation , Signal Transduction , Yohimbine/pharmacology , rap GTP-Binding Proteins/chemistry
2.
Cell Signal ; 22(11): 1681-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600849

ABSTRACT

Three different surface receptors mediate thrombin-induced activation and aggregation of human blood platelets: the protease activated receptors 1 and 4 (PAR1 and PAR4), and the glycoprotein (GP) Ibalpha of the GPIb-IX-V complex. However, their relative contribution in the stimulation of specific intracellular signaling pathways by thrombin remains largely controversial. In this work, we have shown that activation of PAR1 and PAR4 by thrombin or by selective activating peptides stimulated phospholipase C, tyrosine kinases, as well as the small GTPase Rap1b, promoted actin polymerization and cytoskeleton reorganization. When platelets were desensitized for both PAR1 and PAR4, high doses of thrombin, were unable to activate Rap1b, but produced a still evident stimulation of phospholipase C, as documented by the measurement of intracellular Ca(2+) mobilization and protein kinase C activation. These events were abrogated upon proteolysis of GPIbalpha by the metalloproteinase mocarhagin. In PAR1- and PAR4-desensitized platelets, thrombin also induced tyrosine phosphorylation of some substrates, but, surprisingly, this event was largely independent of GPIbalpha binding, as it persisted upon platelet treatment with mocarhagin. Similarly, thrombin-induced actin polymerization and cytoskeleton reorganization were only minimally altered upon PAR1 and PAR4 inactivation and GPIbalpha proteolysis. Interestingly, none of these events were elicited by enzymatically inactive thrombin. Finally we found that GPIbalpha cleavage reduced, but did not abrogate, platelet aggregation in PAR1- and PAR4-desensitized platelets. These results identify a novel pathway for platelet activation operated by thrombin independently of PAR1, PAR4 and GPIbalpha.


Subject(s)
Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Thrombin/pharmacology , Actins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Cytoskeleton/drug effects , Humans , Metalloendopeptidases/pharmacology , Phosphorylation , Platelet Aggregation/drug effects , Type C Phospholipases/metabolism , rap GTP-Binding Proteins/metabolism
3.
J Biol Chem ; 283(43): 28795-805, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18755689

ABSTRACT

Stimulation of G(q)-coupled receptors activates phospholipase C and is supposed to promote both intracellular Ca(2+) mobilization and protein kinase C (PKC) activation. We found that ADP-induced phosphorylation of pleckstrin, the main platelet substrate for PKC, was completely inhibited not only by an antagonist of the G(q)-coupled P2Y1 receptor but also upon blockade of the G(i)-coupled P2Y12 receptor. The role of G(i) on PKC regulation required stimulation of phosphatidylinositol 3-kinase rather than inhibition of adenylyl cyclase. P2Y12 antagonists also inhibited pleckstrin phosphorylation, Rap1b activation, and platelet aggregation induced upon G(q) stimulation by the thromboxane A(2) analogue U46619. Importantly, activation of phospholipase C and intracellular Ca(2+) mobilization occurred normally. Phorbol 12-myristate 13-acetate overcame the inhibitory effect of P2Y12 receptor blockade on PKC activation but not on Rap1b activation and platelet aggregation. By contrast, inhibition of diacylglycerol kinase restored both PKC and Rap1b activity and caused platelet aggregation. Stimulation of P2Y12 receptor or direct inhibition of diacylglycerol kinase potentiated the effect of membrane-permeable sn-1,2-dioctanoylglycerol on platelet aggregation and pleckstrin phosphorylation, in association with inhibition of its phosphorylation to phosphatidic acid. These results reveal a novel and unexpected role of the G(i)-coupled P2Y12 receptor in the regulation of diacylglycerol-mediated events in activated platelets.


Subject(s)
Blood Platelets/metabolism , Diglycerides/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Purinergic P2/metabolism , Signal Transduction , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/chemistry , Blood Proteins/chemistry , Blood Proteins/pharmacology , Diacylglycerol Kinase/metabolism , Enzyme Inhibitors/pharmacology , Humans , Phosphoproteins/chemistry , Phosphoproteins/pharmacology , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase C/metabolism , Receptors, Purinergic P2Y12 , Thromboxane A2/metabolism
4.
Platelets ; 18(2): 135-42, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17365862

ABSTRACT

In this work we have investigated the ability of epinephrine to trigger the release of intracellular Ca2+ in thrombin-desensitized platelets. Addition of thrombin to platelets in the presence of extracellular EGTA caused a rapid and transient release of Ca2+ from intracellular stores and rendered platelets unresponsive to a second addition of the same agonist. Although epinephrine alone had no effect on intracellular Ca2+ mobilization, its addition to thrombin-desensitized platelets was associated to a rapid and evident secondary release of intracellular Ca2+. This effect of epinephrine was not observed when platelets were desensitized with other agonists able to induce phospholipase C activation, including convulxin, U46619, and ADP. Although the platelet receptor for epinephrine is coupled to the Gi family member Gz, no secondary Ca2+ release was seen in thrombin-desensitized platelets upon stimulation of other Gi-coupled receptors, including the P2Y12 receptor and the CXCR4. Addition of hirudin to thrombin-desensitized platelets prevented epinephrine-promoted secondary release of Ca2+, indicating that thrombin, rather than epinephrine itself, is actually responsible for this event as a consequence of thrombin receptors resensitization. Studies with platelets stimulated with specific PAR1- and PAR4- activating peptides proved that neither one of these thrombin receptors were involved in the secondary epinephrine-assisted Ca2+ release. Moreover, we found that thrombin was still able to induce a reduced, but evident release of Ca2+ from internal stores in PAR1- and PAR4-desensitized platelets, which could be followed by a secondary Ca2+ release upon subsequent addition of epinephrine. Importantly, both the primary and the secondary Ca2+ release induced by thrombin and epinephrine in PAR1- and PAR4-desensitized platelets were abrogated upon cleavage of GPIbalpha by the metalloproteinase mocarhagin. These results demonstrate a direct role of thrombin binding to GPIb-IX-V in the mobilization of Ca2+ from intracellular stores, and reveal that epinephrine can restore this process in desensitized platelets, thus prolonging the effect of thrombin stimulation.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Cytosol/metabolism , Epinephrine/physiology , Platelet Activation/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , GTP-Binding Protein alpha Subunits/physiology , Humans , Thrombin/physiology
5.
J Biol Chem ; 280(26): 24386-95, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15863506

ABSTRACT

Binding of thrombopoietin (TPO) to the cMpl receptor on human platelets potentiates aggregation induced by a number of agonists, including ADP. In this work, we found that TPO was able to restore ADP-induced platelet aggregation upon blockade of the G(q)-coupled P2Y1 purinergic receptor but not upon inhibition of the G(i)-coupled P2Y12 receptor. Moreover, TPO triggered platelet aggregation upon co-stimulation of G(z) by epinephrine but not upon co-stimulation of G(q) by the thromboxane analogue U46619. Platelet aggregation induced by TPO and G(i) stimulation was biphasic, and cyclooxygenase inhibitors prevented the second but not the first phase. In contrast to ADP, TPO was unable to induce integrin alpha(IIb)beta(3) activation, as evaluated by binding of both fibrinogen and PAC-1 monoclonal antibody. However, ADP-induced activation of integrin alpha(IIb)beta(3) was blocked by antagonists of the G(q)-coupled P2Y1 receptor but was completely restored by the simultaneous co-stimulation of cMpl receptor by TPO. Inside-out activation of integrin alpha(IIb)beta(3) induced by TPO and G(i) stimulation occurred independently of thromboxane A(2) production and was not mediated by protein kinase C, MAP kinases, or Rho-dependent kinase. Importantly, TPO and G(i) activation of integrin alpha(IIb)beta(3) was suppressed by wortmannin and Ly294002, suggesting a critical regulation by phosphatidylinositol 3-kinase. We found that TPO did not activate phospholipase C in human platelets and was unable to restore ADP-induced phospholipase C activation upon blockade of the G(q)-coupled P2Y1 receptor. TPO induced a rapid and sustained activation of the small GTPase Rap1B through a pathway dependent on phosphatidylinositol 3-kinase. In ADP-stimulated platelets, Rap1B activation was reduced, although not abolished, upon blockade of the P2Y1 receptor. However, accumulation of GTP-bound Rap1B in platelets activated by co-stimulation of cMpl and P2Y12 receptor was identical to that induced by the simultaneous ligation of P2Y1 and P2Y12 receptor by ADP. These results indicate that TPO can integrate G(i), but not G(q), stimulation and can efficiently support integrin alpha(IIb)beta(3) activation platelet aggregation by an alternative signaling pathway independent of phospholipase C but involving the phosphatidylinositol 3-kinase and the small GTPase Rap1B.


Subject(s)
Blood Platelets/cytology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombopoietin/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/chemistry , Androstadienes/pharmacology , Antibodies, Monoclonal/chemistry , Blood Proteins/chemistry , Blood Proteins/metabolism , Calcium/metabolism , Chromones/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Cytosol/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibrinogen/chemistry , Fibrinogen/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Platelet Activation , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, Cytokine/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1 , Receptors, Thrombopoietin , Thrombopoietin/chemistry , Thrombopoietin/metabolism , Thrombopoietin/physiology , Thromboxane A2/metabolism , Thromboxanes/chemistry , Time Factors , Type C Phospholipases/metabolism , Wortmannin , rap GTP-Binding Proteins/metabolism
6.
Blood ; 105(1): 115-21, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15198955

ABSTRACT

The impact of estrogens on the cardiovascular system and their ability to regulate platelet function are matters of controversy. The recent finding that estrogen receptors are expressed in human platelets renders these cells an excellent model for studying the nongenomic effects of these hormones. In this work, we investigated 17beta-estradiol-dependent signaling in platelets from adult healthy men. 17beta-estradiol caused the rapid phosphorylation of the tyrosine kinases Src and Pyk2 and the formation of a signaling complex, which included Src, Pyk2, and the phosphatidylinositol 3-kinase. Both these events were dependent on estrogen receptor beta engagement. We found that estrogen receptor beta was membrane-associated in platelets. On treatment with 17beta-estradiol, Src and Pyk2 activation occurred in the membrane fraction but not in the cytosol. In contrast, no significant activation of phosphatidylinositol 3-kinase was detected in estrogen-treated platelets. 17beta-estradiol did not induce any platelet response directly, but it strongly potentiated the activation of integrin alpha(IIb)beta3 and the platelet aggregation induced by subthreshold concentrations of thrombin. These effects were dependent on estrogen receptor beta recruitment and were associated with a strong synergistic effect with thrombin on Src activation. Taken together, these results indicate that 17beta-estradiol can modulate platelet function by exercising a proaggregating role.


Subject(s)
Blood Platelets/drug effects , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Platelet Aggregation/drug effects , Thrombin/pharmacology , src-Family Kinases/metabolism , Adult , Blood Platelets/cytology , Blood Platelets/enzymology , Blood Platelets/metabolism , Enzyme Activation/drug effects , Focal Adhesion Kinase 2 , Humans , Male , Phosphatidylinositol 3-Kinases/metabolism , Phosphotyrosine/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein-Tyrosine Kinases , Signal Transduction
7.
Exp Cell Res ; 300(1): 84-93, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15383317

ABSTRACT

The expression of the small GTPase Rap1 in human megakaryocytes (MKs) differentiated from cord blood (CB)-derived progenitors was investigated. High levels of Rap1 were detected in the majority of mature megakaryocytes independently of days of culture, while a very low percentage of immature megakaryocytes was found to express a small amount of the protein. Rap1 was predominantly detected on internal alpha-granule but not on the plasma membrane. By contrast, CD41 was clearly present on the peripheral plasma membrane, although it also displayed an intracellular localization similar to that of Rap1. Upon thrombin stimulation, both Rap1 and CD41 translocated to the periphery of the cell. At the opposite, RhoA GTPase and glycoprotein Ibalpha were predominantly located at the plasma membrane and did not undergo relocation upon thrombin stimulation. Thrombin induced a dose- and time-dependent activation of Rap1 in mature megakaryocytes. By using a confocal microscopy approach with a specific probe, active Rap1 was detected exclusively at the peripheral plasma membrane. These results demonstrate that expression of Rap1 occurs during maturation rather than differentiation of megakaryocytes from cord blood progenitor cells. Moreover, we demonstrate that thrombin-activated Rap1 is exclusively localized at the peripheral plasma membrane.


Subject(s)
Cell Membrane/metabolism , Fetal Blood/metabolism , Megakaryocytes/metabolism , rap1 GTP-Binding Proteins/metabolism , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Membrane/drug effects , Cells, Cultured , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Dose-Response Relationship, Drug , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/enzymology , Humans , Infant, Newborn , Megakaryocytes/cytology , Megakaryocytes/drug effects , Megakaryocytes/enzymology , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoprotein IIb/drug effects , Platelet Membrane Glycoprotein IIb/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Thrombin/metabolism , Thrombin/pharmacology , rap1 GTP-Binding Proteins/drug effects , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/metabolism
8.
J Biol Chem ; 279(24): 25299-306, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15078882

ABSTRACT

Thrombin activates human platelets through three different membrane receptors, the protease-activated receptors PAR-1 and PAR-4 and the glycoprotein Ib (GPIb)-IX-V complex. We investigated the contribution of these three receptors to thrombin-induced activation of the small GTPase Rap1B. We found that, similarly to thrombin, selective stimulation of either PAR-1 or PAR-4 by specific activating peptides caused accumulation of GTP-bound Rap1B in a dose-dependent manner. By contrast, in PAR-1- and PAR-4-desensitized platelets, thrombin failed to activate Rap1B. Thrombin, PAR-1-, or PAR-4-activating peptides also induced the increase of intracellular Ca(2+) concentration and the release of serotonin in a dose-dependent manner. We found that activation of Rap1B by selected doses of agonists able to elicit comparable intracellular Ca(2+) increase and serotonin release was differently dependent on secreted ADP. In the presence of the ADP scavengers apyrase or phosphocreatine-phosphocreatine kinase, activation of Rap1B induced by stimulation of either PAR-1 or PAR-4 was totally inhibited. By contrast, thrombin-induced activation of Rap1B was only minimally affected by neutralization of secreted ADP. Concomitant stimulation of both PAR-1 and PAR-4 in the presence of ADP scavengers still resulted in a strongly reduced activation of Rap1B. A similar effect was also observed upon blockade of the P2Y12 receptor for ADP, as well as in P2Y12 receptor-deficient human platelets, but not after blockade of the P2Y1 receptor. Activation of Rap1B induced by thrombin was not affected by preincubation of platelets with the anti-GPIbalpha monoclonal antibody AK2 in the absence of ADP scavengers or a P2Y12 antagonist but was totally abolished when secreted ADP was neutralized or after blockade of the P2Y12 receptor. Similarly, cleavage of the extracellular portion of GPIbalpha by the cobra venom mocarhagin totally prevented Rap1B activation induced by thrombin in the presence of apyrase and in P2Y12 receptor-deficient platelets. By contrast, inhibition of MAP kinases or p160ROCK, which have been shown to be activated upon thrombin binding to GPIb-IX-V, did not affect agonist-induced activation of Rap1B in the presence of ADP scavengers. These results indicate that although both PAR-1 and PAR-4 signal Rap1B activation, the ability of thrombin to activate this GTPase independently of secreted ADP involves costimulation of both receptors as well as binding to GPIb-IX-V.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Receptor, PAR-1/physiology , Receptors, Thrombin/physiology , Thrombin/pharmacology , rap GTP-Binding Proteins/metabolism , Adenosine Diphosphate/physiology , HSP90 Heat-Shock Proteins , Humans , Membrane Proteins/physiology , Platelet Activation , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2Y12
9.
J Biol Chem ; 278(1): 131-8, 2003 Jan 03.
Article in English | MEDLINE | ID: mdl-12407113

ABSTRACT

The small GTP-binding protein Rap1B is activated in human platelets upon stimulation of a G(i)-dependent signaling pathway. In this work, we found that inhibition of platelet adenylyl cyclase by dideoxyadenosine or SQ22536 did not cause activation of Rap1B and did not restore Rap1B activation in platelets stimulated by cross-linking of Fcgamma receptor IIA (FcgammaRIIA) in the presence of ADP scavengers. Moreover, elevation of the intracellular cAMP concentration did not impair the G(i)-dependent activation of Rap1B. Two unrelated inhibitors of phosphatidylinositol 3-kinase (PI3K), wortmannin and LY294002, totally prevented Rap1B activation in platelets stimulated by cross-linking of FcgammaRIIA, by stimulation of the P2Y(12) receptor for ADP, or by epinephrine. However, in platelets from PI3Kgamma-deficient mice, both ADP and epinephrine were still able to normally stimulate Rap1B activation through a PI3K-dependent mechanism, suggesting the involvement of a different isoform of the enzyme. Moreover, the lack of PI3Kgamma did not prevent the ability of epinephrine to potentiate platelet aggregation through a G(i)-dependent pathway. The inhibitory effect of wortmannin on Rap1B activation was overcome by addition of phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)), but not PtdIns(3,4)P(2), although both lipids were found to support phosphorylation of Akt. Moreover, PtdIns(3,4,5)P(3) was able to relieve the inhibitory effect of apyrase on FcgammaRIIA-mediated platelet aggregation. We conclude that stimulation of a G(i)-dependent signaling pathway causes activation of the small GTPase Rap1B through the action of the PI3K product PtdIns(3,4,5)P(3), but not PtdIns(3,4)P(2), and that this process may contribute to potentiation of platelet aggregation.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Blood Platelets/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Serine-Threonine Kinases , Receptors, IgG , Second Messenger Systems/physiology , rap GTP-Binding Proteins/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Adrenergic Agonists/pharmacology , Androstadienes/pharmacology , Animals , Antimetabolites/pharmacology , Apyrase/pharmacology , Blood Platelets/drug effects , Chromones/pharmacology , Dideoxyadenosine/pharmacology , Enzyme Inhibitors/pharmacology , Epinephrine/pharmacology , Humans , Mice , Mice, Knockout , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Platelet Activation/physiology , Platelet Aggregation/physiology , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Fc/metabolism , Wortmannin
10.
Thromb Haemost ; 87(3): 509-17, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11916084

ABSTRACT

Stimulation of human platelets with von Willebrand factor (vWF) induces the rapid tyrosine phosphorylation of several proteins, but very little is known on the tyrosine kinases involved in this process. In the present work, we investigated and compared the activation of two related tyrosine kinases expressed in platelets: the proline-rich tyrosine kinase 2 (Pyk2) and the focal adhesion kinase (FAK). Both kinases were tyrosine phosphorylated upon vWF interaction with glycoprotein Ib-IX-V complex, but with different mechanisms. Tyrosine phosphorylation of FAK was totally dependent on thromboxane A2 production, and was inhibited by the integrin alphaIIbeta3 antagonist RGDS peptide. Moreover, chelation of intracellular calcium or inhibition of protein kinase C (PKC) totally blocked vWF-induced tyrosine phosphorylation of FAK, indicating that this event is downstream phospholipase A2 and phospholipase C activation. By contrast, tyrosine phosphorylation of Pyk2 was only partially reduced by aspirin and RGDS, and was not affected by either calcium chelation or PKC inhibition, suggesting that activation of this kinase does not require phospholipase-mediated signalling. Both FAK and Pyk2 translocated to the cytoskeleton upon vWF stimulation of human platelets by a mechanism depending on agonist-induced actin polymerisation. Prevention of cytoskeletal relocation of Pyk2 and FAK by cytochalasin D totally blocked vWF-induced tyrosine phosphorylation of both kinases. Finally, phosphorylation of Pyk2 induced by vWF, but not by thrombin, was inhibited by piceatannol, suggesting that this kinase lies downstream Syk. These results demonstrate that both Pyk2 and FAK are involved in platelet stimulation by vWF, but indicate that only Pyk2 may play a role in the early signal transduction events activated by ligand binding to glycoprotein Ib-IX-V.


Subject(s)
Platelet Activation/physiology , Protein-Tyrosine Kinases/physiology , von Willebrand Factor/physiology , Blood Platelets/enzymology , Blood Platelets/metabolism , Cytoskeleton/metabolism , Enzyme Activation , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Phosphorylation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Transport , Protein-Tyrosine Kinases/metabolism , Signal Transduction , von Willebrand Factor/metabolism
11.
J Biol Chem ; 277(14): 12009-15, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11815620

ABSTRACT

Stimulation of human platelets by cross-linking of the low affinity receptor for immunoglobulin, FcgammaRIIA, caused the rapid activation of the small GTPase Rap1B, as monitored by accumulation of the GTP-bound form of the protein. This process was totally dependent on the action of secreted ADP since it was completely prevented in the presence of either apyrase or creatine phosphate and creatine phosphokinase. Dose-dependent experiments revealed that the inhibitory effect of ADP scavengers was not related to the reduced increase of cytosolic Ca(2+) concentration in stimulated platelets. Activation of Rap1B induced by clustering of FcgammaRIIA was totally suppressed by AR-C69931MX, a specific antagonist of the G(i)-coupled ADP receptor P2Y12, but was not affected by blockade of the G(q)-coupled receptor, P2Y1. Similarly, direct stimulation of platelets with ADP induced the rapid activation of Rap1B. Pharmacological blockade of the P2Y1 receptor totally prevented ADP-induced Ca(2+) mobilization but did not affect activation of Rap1B. By contrast, prevention of ADP binding to the P2Y12 receptor totally suppressed activation of Rap1B without affecting Ca(2+) signaling. In platelets stimulated by cross-linking of FcgammaRIIA, inhibition of Rap1B activation by ADP scavengers could be overcome by the simultaneous recruitment of the G(i)-coupled alpha(2A)-adrenergic receptor by epinephrine. By contrast, serotonin, which binds to a G(q)-coupled receptor, could not restore activation of Rap1B. When tested alone, epinephrine was found to be able to induce GTP binding to Rap1B, whereas serotonin produced only a slight effect. Finally, activation of Rap1B induced by stimulation of the G(q)-coupled thromboxane A(2) receptor by was completely inhibited by ADP scavengers under conditions in which intracellular Ca(2+) mobilization was unaffected. Inhibition of -induced Rap1B activation was also observed upon blockade of the P2Y12 but not of the P2Y1 receptor for ADP. These results demonstrate that stimulation of a G(i)-dependent signaling pathway by either ADP of epinephrine is necessary and sufficient to activate the small GTPase Rap1B.


Subject(s)
Blood Platelets/enzymology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , rap GTP-Binding Proteins/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Antigens, CD/metabolism , Blood Platelets/metabolism , Calcium/metabolism , Chromatography, Gel , Cross-Linking Reagents/pharmacology , Dimerization , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epinephrine/pharmacology , Humans , Platelet Aggregation Inhibitors/pharmacology , Protein Binding , Receptors, IgG/metabolism , Recombinant Proteins/metabolism , Serotonin/pharmacology , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...