Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 2018 Jun 08.
Article in English | MEDLINE | ID: mdl-29883056

ABSTRACT

Essentials RAS proteins are expressed in platelets but their functions are largely uncharacterized. TC21/RRas2 is required for glycoprotein VI-induced platelet responses and for thrombus stability in vivo. TC21 regulates platelet aggregation by control of αIIb ß3 integrin activation, via crosstalk with Rap1b. This is the first indication of functional importance of a proto-oncogenic RAS protein in platelets. SUMMARY: Background Many RAS family small GTPases are expressed in platelets, including RAC, RHOA, RAP, and HRAS/NRAS/RRAS1, but most of their signaling and cellular functions remain poorly understood. Like RRAS1, TC21/RRAS2 reverses HRAS-induced suppression of integrin activation in CHO cells. However, a role for TC21 in platelets has not been explored. Objectives To determine TC21 expression in platelets, TC21 activation in response to platelet agonists, and roles of TC21 in platelet function in in vitro and in vivo thrombosis. Results We demonstrate that TC21 is expressed in human and murine platelets, and is activated in response to agonists for the glycoprotein (GP) VI-FcRγ immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor, in an Src-dependent manner. GPVI-induced platelet aggregation, integrin αIIb ß3 activation, and α-granule and dense granule secretion, as well as phosphorylation of Syk, phospholipase Cγ2, AKT, and extracellular signal-regulated kinase, were inhibited in TC21-deficient platelets ex vivo. In contrast, these responses were normal in TC21-deficient platelets following stimulation with P2Y, protease-activated receptor 4 and C-type lectin receptor 2 receptor agonists, indicating that the function of TC21 in platelets is GPVI-FcRγ-ITAM-specific. TC21 was required for GPVI-induced activation of Rap1b. TC21-deficient mice did not show a significant delay in injury-induced thrombosis as compared with wild-type controls; however, thrombi were unstable. Hemostatic responses showed similar effects. Conclusions TC21 is essential for GPVI-FcRγ-mediated platelet activation and for thrombus stability in vivo via control of Rap1b and integrins.

2.
J Thromb Haemost ; 16(6): 1211-1225, 2018 06.
Article in English | MEDLINE | ID: mdl-29575487

ABSTRACT

Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY: Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3ß. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Blood Platelets/enzymology , Mitogen-Activated Protein Kinases/blood , Platelet Aggregation/drug effects , Proto-Oncogene Proteins c-raf/blood , Pulmonary Embolism/enzymology , Thrombosis/enzymology , Thromboxanes/blood , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/blood , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Blood Platelets/drug effects , Disease Models, Animal , Humans , Mice, Knockout , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/prevention & control , Pyrimidines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Thrombosis/blood , Thrombosis/genetics , Thrombosis/prevention & control
3.
Thromb Haemost ; 111(3): 508-17, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24352480

ABSTRACT

3-phosphoinositide-dependent protein kinase 1 (PDK1), a member of the protein A,G and C (AGC) family of proteins, is a Ser/Thr protein kinase that can phosphorylate and activate other protein kinases from the AGC family, including Akt at Thr308, all of which play important roles in mediating cellular responses. The functional role of PDK1 or the importance of phosphorylation of Akt on Thr308 for its activity has not been investigated in human platelets. In this study, we tested two pharmacological inhibitors of PDK1, BX795 and BX912, to assess the role of Thr308 phosphorylation on Akt. PAR4-induced phosphorylation of Akt on Thr308 was inhibited by BX795 without affecting phosphorylation of Akt on Ser473. The lack of Thr308 phosphorylation on Akt also led to the inhibition of PAR4-induced phosphorylation of two downstream substrates of Akt, viz. GSK3ß and PRAS40. In vitro kinase activity of Akt was completely abolished if Thr308 on Akt was not phosphorylated. BX795 caused inhibition of 2-MeSADP-induced or collagen-induced aggregation, ATP secretion and thromboxane generation. Primary aggregation induced by 2-MeSADP was also inhibited in the presence of BX795. PDK1 inhibition also resulted in reduced clot retraction indicating its role in outside-in signalling. These results demonstrate that PDK1 selectively phosphorylates Thr308 on Akt thereby regulating its activity and plays a positive regulatory role in platelet physiological responses.


Subject(s)
Blood Platelets/physiology , Oncogene Protein v-akt/metabolism , Threonine/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/metabolism , Blood Platelets/drug effects , Cells, Cultured , Clot Retraction/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Phosphorylation/drug effects , Platelet Activation/drug effects , Pyrimidines/pharmacology , Receptors, Thrombin/metabolism , Serine/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Thromboxanes/metabolism
4.
J Thromb Haemost ; 11(10): 1855-66, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23902231

ABSTRACT

BACKGROUND: 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) is a tumor vascular disrupting agent under clinical trials as an adjacent antitumor agent. DMXAA is structurally similar to flavone-8-acetic acid (FAA), an old tumor vascular disrupting agent with antiplatelet and antithrombotic effects. In contrast to FAA, which causes bleeding in tumor patients, no bleeding has been reported in patients receiving DMXAA. Whether DMXAA also affects platelet function is not clear. OBJECTIVES: To determine the effects of DMXAA on platelet function and explore the underlying mechanisms. METHODS AND RESULTS: DMXAA concentration-dependently inhibited human platelet aggregation and ATP release induced by U46619, arachidonic acid, ADP, collagen, or ristocetin. Furthermore, DMXAA inhibited phosphorylation of Erk1/2 and Akt downstream of thromboxane A2 signaling inhibition. DMXAA also inhibited human platelet phosphodiesterase. The antiplatelet effects were further confirmed using mice administered DMXAA intravenously. DMXAA dramatically inhibited thrombus formation in FeCl3 -injured mouse mesenteric arterial thrombus model and laser-injured mouse cremaster arteriole thrombus model. Notably, at a dose exhibiting antithrombotic effects similar to those of clopidogrel in mice, DMXAA did not significantly increase bleeding. CONCLUSIONS: For the first time, we found that tumor vascular disrupting agent DMXAA has potent antiplatelet and antithrombotic effects without any bleeding diathesis. As DMXAA inhibits platelet activity with safe profile, DMXAA could be used as an efficacious and safe antiplatelet drug.


Subject(s)
Antineoplastic Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Platelet Activation/drug effects , Signal Transduction/drug effects , Thrombosis/prevention & control , Thromboxane A2/antagonists & inhibitors , Xanthones/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Triphosphate/metabolism , Animals , Humans , MAP Kinase Signaling System , Mice , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Thromboxane A2/metabolism , Thromboxane-A Synthase/metabolism
5.
J Thromb Haemost ; 11 Suppl 1: 340-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23809137

ABSTRACT

Recent work by the Encyclopedia of DNA Elements project showed that non-protein-coding RNAs account for an unexpectedly large proportion of the human genome. Among these non-coding RNAs are microRNAs (miRNAs), which are small RNA molecules that modulate protein expression by degrading mRNA or repressing mRNA translation. MiRNAs have been shown to play important roles in hematopoiesis including embryonic stem cell differentiation, erythropoiesis, granulocytopoiesis/monocytopoiesis, lymphopoiesis, and megakaryocytopoiesis. Additionally, disordered miRNA biogenesis and quantitative or qualitative alterations in miRNAs and their targets are associated with hematological pathologies. Platelets contain machinery to process pre-miRNAs into mature miRNAs, and specific platelet miRNA levels have been found to correlate with platelet reactivity. This review summarizes the current state of knowledge of miRNAs in megakaryocytes and platelets, and the exciting possibilities for future megakaryocyte-platelet transcriptome research.


Subject(s)
Blood Platelets/cytology , MicroRNAs/metabolism , Platelet Activation , Blood Platelets/metabolism , Humans
6.
J Thromb Haemost ; 11(1): 149-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23140172

ABSTRACT

BACKGROUND: Platelet ADP receptor P2Y(12) is well studied and recognized as a key player in platelet activation, hemostasis and thrombosis. However, the role of P2Y(12) in platelet apoptosis remains unknown. OBJECTIVES: To evaluate the role of the P2Y(12) receptor in platelet apoptosis. METHODS: We used flow cytometry and Western blotting to assess apoptotic events in platelets treated with ABT-737 or ABT-263, and stored at 37°C, combined with P2Y(12) receptor antagonists or P2Y(12) -deficient mice. RESULTS: P2Y(12) activation attenuated apoptosis induced by ABT-737 in human and mouse platelets in vitro, evidenced by reduced phosphatidylserine (PS) exposure, diminished depolarization of mitochondrial inner transmembrane potential (ΔΨm) and decreased caspase-3 activation. Through increasing the phosphorylation level of Akt and Bad, and changing the interaction between different Bcl-2 family proteins, P2Y(12) activation inactivated Bak/Bax. This antiapoptotic effect could be abolished by P2Y(12) antagonism or PI3K inhibition. We also observed the antiapoptotic effect of P2Y(12) activation in platelets stored at 37°C. P2Y(12) activation improved the impaired activation responses of apoptotic platelets stressed by ABT-737. In platelets from mice dosed with ABT-263 in vivo, clopidogrel or deficiency of P2Y(12) receptor enhanced apoptosis along with increased Bak/Bax activation. CONCLUSIONS: This study demonstrates that P2Y(12) activation protects platelets from apoptosis via PI3k-dependent Bak/Bax inactivation, which may be physiologically important to counter the proapoptotic challenge. Our findings that P2Y(12) blockade exaggerates platelet apoptosis induced by ABT-263 (Navitoclax) also imply a novel drug interaction of ABT-263 and P2Y(12) antagonists.


Subject(s)
Apoptosis , Blood Platelets/drug effects , Blood Platelets/enzymology , Phosphatidylinositol 3-Kinase/blood , Receptors, Purinergic P2Y12/blood , bcl-2 Homologous Antagonist-Killer Protein/blood , bcl-2-Associated X Protein/blood , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Blood Platelets/pathology , Blotting, Western , Caspase 3/blood , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrophenols/pharmacology , Phosphatidylserines/blood , Phosphorylation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/blood , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/deficiency , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/genetics , Signal Transduction , Sulfonamides/pharmacology , Time Factors , bcl-Associated Death Protein/blood
7.
J Thromb Haemost ; 10(10): 2149-57, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22906019

ABSTRACT

BACKGROUND: In our previous in vitro study, we reported a constitutively active chimeric P2Y(12) (cP2Y(12)) and found that AR-C78511 is a potent inverse agonist at this receptor. The role of cP2Y(12) in platelet activation and thrombosis is not clear. OBJECTIVES: To investigate the physiologic implications of cP2Y(12) for platelet activation and thrombus formation, and to evaluate the antiplatelet activity of AR-C78511 as an inverse agonist. METHODS AND RESULTS: We generated transgenic mice conditionally and platelet-specifically expressing cP2Y(12). High-level expression of cP2Y(12) in platelets increased platelet reactivity, as shown by increased platelet aggregation in response to multiple platelet agonists. Moreover, transgenic mice showed a shortened bleeding time, and more rapid and stable thrombus formation in mesenteric artery injured with FeCl(3). The constitutive activity of cP2Y(12) in platelets was confirmed by decreased platelet cAMP levels and constitutive Akt phosphorylation in the absence of agonists. AR-C78511 reversed the cAMP decrease in transgenic mouse platelets, and exhibited a superior antiplatelet effect to that of AR-C69931MX in transgenic mice. CONCLUSIONS: These findings further emphasize the importance of P2Y(12) in platelet activation, hemostasis, and thrombosis, as well as the prothrombotic role of the constitutive activity of P2Y(12). Our data also validate the in vivo inverse agonist activity of AR-C78511, and confirm its superior antiplatelet activity over neutral antagonists.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Receptors, Purinergic P2Y12/metabolism , Thrombosis/blood , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Animals , Bleeding Time , Blood Platelets/drug effects , Chlorides , Cyclic AMP/blood , Disease Models, Animal , Drug Inverse Agonism , Ferric Compounds , Fibrinolytic Agents/pharmacology , Genotype , Mice , Mice, Transgenic , Phenotype , Phosphorylation , Platelet Activation/drug effects , Platelet Activation/genetics , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/blood , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/genetics , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/prevention & control
8.
J Thromb Haemost ; 9(4): 810-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21261805

ABSTRACT

BACKGROUND: In hypercholesterolemia, platelets demonstrate increased reactivity and promote the development of cardiovascular disease. OBJECTIVE: This study was carried out to investigate the contribution of the ADP receptor P2Y12-mediated pathway to platelet hyperreactivity due to hypercholesterolemia. METHODS: Low-density lipoprotein receptor-deficient mice and C57Bl/6 wild-type mice were fed on normal chow and high-fat (Western or Paigen) diets for 8 weeks to generate differently elevated cholesterol levels. P2Y12 receptor-induced functional responses via G(i) signaling were studied ex vivo when washed murine platelets were activated by 2MeSADP and PAR4 agonist AYPGKF in the presence and absence of indomethacin. Platelet aggregation and secretion, α(IIb)ß(3) receptor activation and the phosphorylation of extracellular signal-regulated protein kinase (ERK) and Akt were analyzed. RESULTS: Plasma cholesterol levels ranged from 69 ± 10 to 1011 ± 185 mg dL(-1) depending on diet in mice with different genotypes. Agonist-dependent aggregation, dense and α-granule secretion and JON/A binding were gradually and significantly (P < 0.05) augmented at low agonist concentration in correlation with the increasing plasma cholesterol levels, even if elevated thromboxane generation was blocked. These functional responses were induced via increased levels of G(i) -mediated ERK and Akt phosphorylation in hypercholesterolemic mice vs. normocholesterolemic animals. In addition, blocking of the P2Y12 receptor by AR-C69931MX (Cangrelor) resulted in strongly reduced platelet aggregation in mice with elevated cholesterol levels compared with normocholesterolemic controls. CONCLUSIONS: These data revealed that the P2Y12 receptor pathway was substantially involved in platelet hyperreactivity associated with mild and severe hypercholesterolemia.


Subject(s)
Blood Platelets/pathology , Hypercholesterolemia/pathology , Receptors, Purinergic P2Y12/physiology , Animals , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
10.
J Thromb Haemost ; 8(10): 2283-93, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20670370

ABSTRACT

BACKGROUND: Myosin IIA is an essential platelet contractile protein that is regulated by phosphorylation of its regulatory light chain (MLC) on residues (Thr)18 and (Ser)19 via the myosin light chain kinase (MLCK). OBJECTIVE: The present study was carried out to elucidate the mechanisms regulating MLC (Ser)19 and (Thr)18 phosphorylation and the functional consequence of each phosphorylation event in platelets. RESULTS: Induction of 2MeSADP-induced shape change occurs within 5s along with robust phosphorylation of MLC (Ser)19 with minimal phosphorylation of MLC (Thr)18. Selective activation of G(12/13) produces both slow shape change and comparably slow MLC (Thr)18 and (Ser)19 phosphorylation. Stimulation with agonists that trigger ATP secretion caused rapid MLC (Ser)19 phosphorylation while MLC (Thr)18 phosphorylation was coincident with secretion. Platelets treated with p160(ROCK) inhibitor Y-27632 exhibited a partial inhibition in secretion and had a substantial inhibition in MLC (Thr)18 phosphorylation without effecting MLC (Ser)19 phosphorylation. These data suggest that phosphorylation of MLC (Ser)19 is downstream of Gq/Ca(2+) -dependent mechanisms and sufficient for shape change, whereas MLC (Thr)18 phosphorylation is substantially downstream of G(12/13) -regulated Rho kinase pathways and necessary, probably in concert with MLC (Ser)19 phosphorylation, for full contractile activity leading to dense granule secretion. Overall, we suggest that the amplitude of the platelet contractile response is differentially regulated by a least two different signaling pathways, which lead to different phosphorylation patterns of the myosin light chain, and this mechanism results in a graded response rather than a simple on/off switch.


Subject(s)
Blood Platelets/metabolism , Myosin Light Chains/chemistry , Myosin Light Chains/genetics , Serine/chemistry , Threonine/chemistry , Adenosine Diphosphate/chemistry , Calcium/chemistry , Calcium/metabolism , Cell Shape , Humans , Kinetics , Phosphorylation , Platelet Aggregation , Signal Transduction , rho-Associated Kinases/metabolism
11.
J Thromb Haemost ; 5(2): 362-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17096705

ABSTRACT

BACKGROUND: It has been reported that the association of glycoprotein VI (GPVI) with lipid rafts regulates GPVI signaling in platelets. OBJECTIVE: Secreted adenosine 5'-diphosphate (ADP) potentiates GPVI-induced platelet aggregation at particular agonist concentrations. We have investigated whether the decrease in GPVI signaling, previously reported in platelets with disrupted rafts, is a result of the loss of agonist potentiation by ADP. METHODS: We disrupted platelet lipid rafts with methyl-beta-cyclodextrin and measured signaling events downstream of GPVI activation. RESULTS: Lipid raft disruption decreases aggregation induced by low concentrations of convulxin, but this decrease is almost eliminated in the presence of ADP antagonists. Signaling indicators, such as protein phosphorylation and calcium mobilization, were not affected by raft disruption in collagen or convulxin stimulated platelets. Interestingly, however, raft disruption directly reduced GPVI signaling induced by collagen-related peptide. CONCLUSIONS: Lipid rafts do not directly contribute to signaling by the physiologic agonist collagen. The effects of disruption of lipid rafts in in vitro assays can be attributed to inhibition of ADP feedback that potentiates GPVI signaling.


Subject(s)
Membrane Microdomains/physiology , Platelet Membrane Glycoproteins/agonists , Adenosine Diphosphate/antagonists & inhibitors , Blood Platelets , Collagen/pharmacology , Feedback, Physiological , Humans , Platelet Aggregation , Signal Transduction
12.
Curr Drug Targets ; 7(10): 1253-63, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17073586

ABSTRACT

Platelet activation and aggregation is an integral component of the pathophysiology that leads to thrombotic and ischemic diseases such as cerebral stroke, peripheral vascular disease and myocardial infarction. Anti-platelet agents (such as aspirin, ADP receptor antagonists, and GPIIb/IIIa antagonists), phosphodiesterase inhibitors and anti-coagulants are major part of the current treatment towards treating ischemic diseases. However, their limited efficacy in the setting of arterial thrombosis, unfavorable side effect profile and cost-to-benefit issues substantiate the need for the development of newer and more efficacious antithrombotic drugs. Various platelet agonists like adenosine diphosphate (ADP), thrombin and thromboxane A2 (TXA2) activate platelets by acting via their respective surface receptors, which couple to one or more distinct G-proteins belonging to either the G(i), G(q), G(12/13) or G(s) families. Upon activation, each of these G-proteins trigger a series of intracellular signaling cascades, causing the platelets to undergo shape change, secrete their granular contents, generate positive feedback mediators and form stable platelet aggregates. In addition, various G-protein-mediated signaling cascades act in synergy with one another to amplify the magnitude of the platelet responses. The significance of G-proteins as key mediators of the platelet function and normal hemostasis is further corroborated by extensive gene knockout studies. In this review we will limit our discussion to understanding the role of G-proteins in the process of platelet activation and discuss some of the anti-thrombotic drugs that mediate their beneficial effects by interfering with or preventing the initiation of the G-protein signaling pathway.


Subject(s)
Blood Platelets/physiology , GTP-Binding Proteins/blood , Platelet Activation/physiology , Signal Transduction/physiology , Animals , Blood Platelets/drug effects , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/physiology , Humans , Platelet Activation/drug effects , Signal Transduction/drug effects
13.
J Thromb Haemost ; 4(11): 2317-26, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17059469

ABSTRACT

Upon injury to a vessel wall the exposure of subendothelial collagen results in the activation of platelets. Platelet activation culminates in shape change, aggregation, release of granule contents and generation of lipid mediators. These secreted and generated mediators trigger a positive feedback mechanism potentiating the platelet activation induced by physiological agonists such as collagen and thrombin. Adenine nucleotides, adenosine diphosphate (ADP) and adenosine triphosphate (ATP), released from damaged cells and that are secreted from platelet-dense granules, contribute to the positive feedback mechanism by acting through nucleotide receptors on the platelet surface. ADP acts through two G protein-coupled receptors, the Gq-coupled P2Y1 receptor, and the Gi-coupled P2Y12 receptor. ATP, on the other hand, acts through the ligand-gated channel P2X1. Stimulation of platelets by ADP leads to shape change, aggregation and thromboxane A2 generation. ADP-induced dense granule release depends on generated thromboxane A2. Furthermore, costimulation of both P2Y1 and P2Y12 receptors is required for ADP-induced platelet aggregation. ATP stimulation of P2X1 is involved in platelet shape change and helps to amplify platelet responses mediated by agonists such as collagen. Activation of each of these nucleotide receptors results in unique signal transduction pathways that are important in the regulation of thrombosis and hemostasis.


Subject(s)
Adenine Nucleotides/metabolism , Blood Platelets/metabolism , Platelet Aggregation , Receptors, Purinergic/metabolism , Signal Transduction , Animals , Blood Vessels/injuries , Blood Vessels/metabolism , Collagen/metabolism , Cytoplasmic Granules/metabolism , Humans , Purinergic Agonists , Thrombin/metabolism , Thromboxane A2/metabolism
14.
J Thromb Haemost ; 4(12): 2678-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16995904

ABSTRACT

BACKGROUND: Galphaq (Gene GNAQ) plays a major role in platelet signal transduction but little is known regarding its transcriptional regulation. OBJECTIVES: We studied Galphaq promoter activity using luciferase reporter gene assays in human erythroleukemia (HEL) cells treated with phorbol 12-myristate 13-acetate (PMA) for 24 h to induce megakaryocytic transformation. METHODS AND RESULTS: PMA-treated HEL cells showed enhanced Galphaq expression. Reporter (luciferase) gene studies on 5' upstream construct (up to -116 bp from ATG) revealed a negative regulatory site at -238/-202 and two positive sites at -203/-138 and -1116/-731. The positive regulatory region -203/-138 contained overlapping Sp1/AP-2/EGR-1 consensus sites. Gel shift studies on Galphaq oligonucleotides 1 (-203/-175) and 2 (-174/-152) using HEL cell extracts demonstrated protein binding that was due to early growth response factor EGR-1 at two sites. Mutations in either EGR-1 site markedly decreased the gene activity, indicating functional relevance. Mutation of consensus E-Box motif (-185/-180) had no effect. Reduction in the expression of endogenous EGR-1 with antisense oligonucleotide to EGR-1 inhibited PMA-induced Galphaq transcription. Correspondingly, Egr-1 deficient mouse platelets also showed approximately 50% reduction in the Galphaq expression relative to wild-type platelets. CONCLUSIONS: These studies suggest that Galphaq gene is regulated during PMA-induced megakaryocytic differentiation by EGR-1, an early growth response transcription factor that regulates a wide array of genes and plays a major role in diverse activities, including cell proliferation, differentiation and apoptosis, and in vascular response to injury and atherosclerosis.


Subject(s)
Early Growth Response Protein 1/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Megakaryocytes/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Blood Platelets/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Early Growth Response Protein 1/genetics , Electrophoretic Mobility Shift Assay , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Deletion , Genes, Reporter , Humans , Luciferases , Luminescent Agents , Megakaryocytes/cytology , Megakaryocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Oligonucleotides/genetics , Oligonucleotides/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Transfection
15.
J Thromb Haemost ; 4(3): 638-47, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16460446

ABSTRACT

BACKGROUND: Thromboxane A2 (TXA2) is a positive feedback lipid mediator that is generated upon stimulation of platelets with various agonists. Aspirin works as an antithrombotic drug by blocking the generation of TXA2. The aim of this study was to evaluate the role of the purinergic P2Y receptors in thrombin-induced TXA2 generation. RESULTS: PAR1-activating peptide (SFLLRN), PAR4-activating peptide (AYPGKF), and thrombin, induced the activation of cytosolic phospholipase A2 (cPLA2), release of arachidonic acid (AA) from membrane-bound phospholipids, and subsequent TXA2 generation in human platelets. The actions of these agonists were significantly inhibited in the presence of the P2Y12 receptor antagonist, AR-C69931MX, but not the P2Y1 receptor antagonist, MRS2179. In addition, AYPGKF- and thrombin-induced TXA2 generation was significantly reduced in platelets from mice dosed with clopidogrel, confirming the results obtained with the human platelets. Also, Pearl mouse platelets that lack releasable nucleotides generated significantly less TXA2 when compared with the wild-type littermates in response to PAR stimulation. Inhibition of extracellular signal-regulated protein kinase 1/2 (Erk 1/2) activation using U0126, an inhibitor of MAP kinase kinase (MEK), suppressed PAR-mediated cPLA2 phosphorylation and TXA2 generation. Further, platelets that were pretreated with AR-C69931MX, as well as Pearl mouse platelets, displayed the reduced levels of Erk1/2 phosphorylation upon stimulation with the PAR agonists. CONCLUSIONS: Based on these findings, we conclude that thrombin-induced Erk1/2 activation is essential for PAR-mediated TXA2 generation, which is potentiated by the P2Y12 receptor-mediated signaling pathway but not the P2Y1 receptor-mediated signaling pathway. Finally, using selective inhibitors of Src kinases, we show that PAR-mediated Src activation precedes Erk1/2 activation.


Subject(s)
Blood Platelets/drug effects , Membrane Proteins/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Purinergic P2/drug effects , Thrombin/pharmacology , Thromboxane A2/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Animals , Blood Platelets/enzymology , Butadienes/pharmacology , Clopidogrel , Enzyme Inhibitors/pharmacology , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Models, Biological , Nitriles/pharmacology , Phospholipases A/metabolism , Phospholipases A2 , Platelet Aggregation Inhibitors/pharmacology , Pyrimidines , Receptor, PAR-1/drug effects , Receptors, Purinergic P2Y12 , Receptors, Thrombin/drug effects , Signal Transduction , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology , src-Family Kinases/antagonists & inhibitors
16.
J Thromb Haemost ; 3(5): 1036-41, 2005 May.
Article in English | MEDLINE | ID: mdl-15869601

ABSTRACT

ADP is important in propagating hemostasis upon its secretion from activated platelets in response to other agonists. Lipid rafts are microdomains within the plasma membrane that are rich in cholesterol and sphingolipids, and have been implicated in the stimulatory mechanisms of platelet agonists. We sought to determine the importance of lipid rafts in ADP-mediated platelet activation via the G protein-coupled P2Y1 and P2Y12 receptors using lipid raft disruption by cholesterol depletion with methyl-beta-cyclodextrin. Stimulation of cholesterol-depleted platelets with ADP resulted in a reduction in the extent of aggregation but no difference in the extent of shape change or intracellular calcium release. Furthermore, repletion of cholesterol to previously depleted membranes restored ADP-mediated platelet aggregation. In addition, P2Y12-mediated inhibition of cAMP formation was significantly decreased upon cholesterol depletion from platelets. Stimulation of cholesterol-depleted platelets with agonists that depend upon Galpha(i) activation for full activation displayed significant loss of aggregation and secretion, but showed restoration when simultaneously stimulated with the Galpha(z)-coupled agonist epinephrine. Finally, Galpha(i) preferentially localizes to lipid rafts as determined by sucrose density centrifugation. We conclude that Galpha(i) signaling downstream of P2Y12 activation, but not Galpha(q) or Galpha(z) signaling downstream of P2Y1 or alpha2A activation, respectively, has a requirement for lipid rafts that is necessary for its function in ADP-mediated platelet activation.


Subject(s)
Adenosine Diphosphate/metabolism , Blood Platelets/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Membrane Microdomains/chemistry , Membrane Proteins/metabolism , Platelet Activation , Receptors, Purinergic P2/metabolism , Adenosine Diphosphate/chemistry , Blotting, Western , Calcium/metabolism , Centrifugation, Density Gradient , Cholesterol/chemistry , Cholesterol/metabolism , Cyclic AMP/metabolism , Epinephrine/metabolism , Humans , Membrane Microdomains/metabolism , Receptors, Purinergic P2Y12 , Signal Transduction , Sucrose/pharmacology , Time Factors , beta-Cyclodextrins/metabolism
17.
Rev Med Chir Soc Med Nat Iasi ; 109(1): 191-9, 2005.
Article in Romanian | MEDLINE | ID: mdl-16607854

ABSTRACT

Nucleotides are important extracellular signaling molecules. It has been established that nucleotides are released from damaged cells, activated platelets and endothelial cells. Thus, at the site of vascular injury, the concentrations of extracellular nucleotides can become elevated. Nucleotides have been shown to cause mobilization of intracellular calcium, upregulation of Mac-1 (CD11b/CD18), degranulation, and chemotaxis in human neutrophils. The goal of this work is to investigate the functional characteristics of nucleotide-receptors in human neutrophils. Nucleotides (ATP and UTP), caused intracellular calcium mobilization in a dose dependent manner. Pharmacological characterization using selective agonists (ATP, UTP), pertussis toxin in human neutrophils and human astrocytoma cells 1321N1 stably expressing P2Y2 or P2Y4 receptors, revealed that human neutrophils express only functional P2Y2 receptors. Treatment of neutrophils with pertussis toxin causes a partial inhibition of nucleotide-induced calcium mobilization. Similarly, by using 1321N astrocytoma cells expressing the P2Y2 receptor we confirmed that calcium mobilization is only partially inhibited by pertussis toxin. The partial resistance of P2Y2-mediated intracellular calcium mobilization suggests that this receptor subtype is coupled not only to a Gi protein, but also to a protein belonging to the Gq-family (most likely G16). In conclusion, we have shown that human neutrophils express functional P2Y2 receptors and all the nucleotide responses are mediated by P2Y2 receptor subtype and that P2Y2 receptors are the functional able to trigger intracellular signaling event in human neutrophils through dual activation of different G proteins.


Subject(s)
Neutrophils/physiology , Nucleotides/physiology , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/metabolism , Calcium/metabolism , Humans , Neutrophils/metabolism , Nucleotides/metabolism , Receptors, Purinergic P2Y2 , Signal Transduction , Uridine Triphosphate/metabolism
18.
J Thromb Haemost ; 2(6): 978-84, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140134

ABSTRACT

Alpha granule release plays an important role in propagating a hemostatic response upon platelet activation. We evaluated the ability of various agonists to cause alpha granule release in platelets. Alpha granule release was measured by determining P-selectin surface expression in aspirin-treated washed platelets. ADP-induced P-selectin expression was inhibited both by MRS 2179 (a P2Y1 selective antagonist) and AR-C69931MX (a P2Y12 selective antagonist), suggesting a role for both Galpha(q) and Galpha(i) pathways in ADP-mediated alpha granule release. Consistent with these observations, the combination of serotonin (a Galpha(q) pathway stimulator) and epinephrine (a Galpha(z) pathway stimulator) also caused alpha granule release. Furthermore, U46619-induced P-selectin expression was unaffected by MRS 2179 but was dramatically inhibited by AR-C69931, indicating a dominant role for P2Y12 in U46619-mediated alpha granule release. Additionally, the Galpha(12/13)-stimulating peptide YFLLRNP potentiated alpha granule secretion in combination with either ADP or serotonin/epinephrine costimulation but was unable to induce secretion by itself. Finally, costimulation of the Galpha(i) and Galpha(12/13) pathways resulted in a significant dose-dependent increase in alpha granule release. We conclude that ADP-induced alpha granule release in aspirin-treated platelets occurs through costimulation of Galpha(q) and Galpha(i) signaling pathways. The P2Y12 receptor plays an important role in thromboxane A(2)-mediated alpha granule release, and furthermore activation of Galpha(12/13) and Galpha(q) signaling pathway can cause alpha granule release.


Subject(s)
Blood Platelets/metabolism , Receptors, G-Protein-Coupled/physiology , Secretory Vesicles/metabolism , Signal Transduction , Adenosine Diphosphate/pharmacology , Cytoplasmic Granules/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Humans , Membrane Proteins/physiology , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , Thromboxane A2/pharmacology
19.
J Thromb Haemost ; 2(5): 804-12, 2004 May.
Article in English | MEDLINE | ID: mdl-15099288

ABSTRACT

The activated platelet surface serves as an integral part of the prothrombinase complex upon activation by potent platelet agonists such as thrombin and collagen. We determined the receptor specificity through which thrombin was enhancing collagen-induced thrombin generation. Whereas SFLLRN or AYPGKF alone produced minimal thrombin generation or phosphatidylserine exposure through protease activated receptor (PAR) stimulation, they caused a leftward shift in the collagen-induced thrombin generation dose-response curve. Although SFLLRN or AYPGKF potentiated collagen-induced thrombin generation, neither of them potentiated to the same extent as thrombin. However, SFLLRN and AYPGKF together potentiated collagen-induced thrombin generation to the same extent as thrombin. We conclude that thrombin mediates its procoagulant activity through activation of both PAR1 and PAR4 receptors. Similarly, neither PAR1 nor PAR4 stimulation alone mimicked the annexin V-binding response caused by thrombin stimulation. The combination of PAR activating peptides caused minimal increases in annexin V binding, but caused significant thrombin generation, suggesting that events other than phosphatidylserine exposure may play a role in platelet prothrombinase complex formation. We also investigated the ability of ADP to potentiate agonist-induced thrombin generation. Whereas P2Y(1) antagonism did not affect collagen or thrombin-induced thrombin generation, P2Y(12) antagonism did decrease both collagen- and thrombin-induced thrombin generation, suggesting that ADP potentiates thrombin generation primarily through the P2Y(12) receptor. Collectively, these results suggest that stimulation of both the PAR1 and PAR4 receptors are necessary for thrombin-induced procoagulant activity, and that the P2Y(12) receptor, but not the P2Y(1) receptor, is responsible for the potentiation of agonist-induced platelet procoagulant activity.


Subject(s)
Blood Platelets/metabolism , Receptors, Proteinase-Activated/physiology , Receptors, Purinergic P2/physiology , Thrombin/biosynthesis , Annexin A5/metabolism , Collagen/pharmacology , Dose-Response Relationship, Drug , Humans , Membrane Proteins/metabolism , Receptor, PAR-1/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , Receptors, Thrombin/metabolism , Thromboplastin/metabolism
20.
J Thromb Haemost ; 1(4): 814-20, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12871420

ABSTRACT

The integrin alphavbeta3 mediates platelet adhesion to the matrix protein osteopontin and likely is the predominant integrin mediating platelet adhesion to the matrix protein vitronectin. To address the mechanism that regulates alphavbeta3 activity in platelets, we measured the effect of the P2Y1 antagonist adenosine 3'-phosphate-5'-phosphate (A3P5P) and the P2Y12 antagonist AR-C66096 on ADP-stimulated platelet adhesion to osteopontin and vitronectin. Each antagonist completely inhibited platelet adhesion, implying that concurrent stimulation of P2Y1 and P2Y12 was required to activate alphavbeta3. The reducing agent dithiothreitol and Mn2+ also induced platelet adhesion to osteopontin, but did so without stimulating platelet activation. Thus, these data suggest that ADP stimulation regulates alphavbeta3 activity by perturbing the conformation of its extracellular domain. The actin polymerization inhibitors cytochalasin D and latrunculin A also induced platelet adhesion to osteopontin and vitronectin. Thus, alphavbeta3 activity in resting platelets appears to be constrained by the platelet cytoskeleton. Moreover, the effect of these agents was inhibited by A3P5P and AR-C66096 at micromolar and subnanomolar concentrations, respectively, suggesting that subthreshold platelet stimulation by ADP was required. Our data suggest that signals from both Galphaq- and Galphai-coupled receptors converge to release cytoskeletal constraints on alphavbeta3. We propose that the release of cytoskeletal constraints and a concurrent increase in affinity for ligands is responsible for alphavbeta3-mediated platelet adhesion.


Subject(s)
Blood Platelets/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Integrin alphaVbeta3/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Actins/metabolism , Cytoskeleton/metabolism , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gq-G11 , Humans , Osteopontin , Sialoglycoproteins/metabolism , Vitronectin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...