201
|
Reséndiz JC, Kroll MH, Lassila R. Protease-activated receptor-induced Akt activation--regulation and possible function. J Thromb Haemost 2007; 5:2484-93. [PMID: 17883592 DOI: 10.1111/j.1538-7836.2007.02769.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Thrombin induces the activation of the platelet serine/threonine kinase Akt. Akt activation is dependent on its phosphorylation at Thr308 and Ser473. The mechanism by which thrombin induces Akt phosphorylation is controversial, as is the role of Akt in platelet function. OBJECTIVES To investigate how protease-activated receptors (PARs) stimulate Akt and the role that Akt plays in human platelet function. METHODS Platelets were stimulated through PAR1 or PAR4. Specific inhibitors were used to evaluate, by Western blotting, signaling pathways regulating Akt phosphorylation, and the role of activated Akt was evaluated by aggregometry and flow cytometry. RESULTS Phospholipase C (PLC) controls Akt phosphorylation elicited by PARs. Stimulation of PAR1 or PAR4 resulted in rapid Akt phosphorylation, independently of secreted ADP and phosphatidylinositol-3-kinase (PI3K) activation. Akt phosphorylation approximately 60 s after PAR1 stimulation became entirely dependent on the purinergic receptor P2Y(12) and the activation of PI3K. In contrast, PAR4 partially sustained Akt phosphorylation independently of P2Y(12) and PI3K for up to 300 s. Pharmacologic inhibition of Akt reduced P-selectin expression and fibrinogen binding in platelets stimulated through PAR1, and delayed platelet aggregation in response to submaximal PAR1 or PAR4 stimulation, although aggregation at 300 s was unaffected. CONCLUSIONS Platelet PAR stimulation causes rapid Akt phosphorylation downstream of PLC, whereas with continuous stimulation, ADP and PI3K are required for maintaining Akt phosphorylation. Activated Akt regulates platelet function by modulating secretion and alpha(IIb)beta(3) activation.
Collapse
Affiliation(s)
- J C Reséndiz
- Wihuri Research Institute, Kalliolinnantie 4, Helsinki, Finland.
| | | | | |
Collapse
|
202
|
Hassan AE, Zacharatos H, Fareed M, Suri K, Qureshi AI. Drug evaluation of clopidogrel in patients with ischemic stroke. Expert Opin Pharmacother 2007; 8:2825-38. [DOI: 10.1517/14656566.8.16.2825] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Ameer E Hassan
- University of Minnesota, Zeenat Qureshi Stroke Research Center, Department of Neurology, 82 – 100 Phillips Wangensteen Building, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USA
| | - Haralabos Zacharatos
- University of Minnesota, Zeenat Qureshi Stroke Research Center, Department of Neurology, 82 – 100 Phillips Wangensteen Building, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USA
| | - M Fareed
- University of Minnesota, Zeenat Qureshi Stroke Research Center, Department of Neurology, 82 – 100 Phillips Wangensteen Building, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USA
| | - K Suri
- University of Minnesota, Zeenat Qureshi Stroke Research Center, Department of Neurology, 82 – 100 Phillips Wangensteen Building, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USA
| | - Adnan I Qureshi
- University of Minnesota, Zeenat Qureshi Stroke Research Center, Department of Neurology, 82 – 100 Phillips Wangensteen Building, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
203
|
Jones S, Tucker KL, Sage T, Kaiser WJ, Barrett NE, Lowry PJ, Zimmer A, Hunt SP, Emerson M, Gibbins JM. Peripheral tachykinins and the neurokinin receptor NK1 are required for platelet thrombus formation. Blood 2007; 111:605-12. [PMID: 17895403 PMCID: PMC2575837 DOI: 10.1182/blood-2007-07-103424] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Platelets play an important role in hemostasis, with inappropriate platelet activation being a major contributor to debilitating and often fatal thrombosis by causing myocardial infarction and stroke. Although current antithrombotic treatment is generally well tolerated and effective, many patients still experience cardiovascular problems, which may reflect the existence of alternative underlying regulatory mechanisms in platelets to those targeted by existing drugs. In this study, we define a role for peripherally distributed members of the tachykinin family of peptides, namely substance P and the newly discovered endokinins A and B that are present in platelets, in the activation of platelet function and thrombus formation. We have reported previously that the preferred pharmacologically characterized receptor for these peptides, the NK1 receptor, is present on platelets. Inhibition or deficiency of the NK1 receptor, or SP agonist activity, resulted in substantially reduced thrombus formation in vitro under arterial flow conditions, increased bleeding time in mice, and a decrease in experimentally induced thromboembolism. Inhibition of the NK1 receptor may therefore provide benefit in patients vulnerable to thrombosis and may offer an alternative therapeutic target.
Collapse
Affiliation(s)
- Sarah Jones
- School of Biological Sciences, University of Reading, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Erlinge D, Burnstock G. P2 receptors in cardiovascular regulation and disease. Purinergic Signal 2007; 4:1-20. [PMID: 18368530 PMCID: PMC2245998 DOI: 10.1007/s11302-007-9078-7] [Citation(s) in RCA: 280] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Accepted: 08/22/2007] [Indexed: 12/11/2022] Open
Abstract
The role of ATP as an extracellular signalling molecule is now well established and evidence is accumulating that ATP and other nucleotides (ADP, UTP and UDP) play important roles in cardiovascular physiology and pathophysiology, acting via P2X (ion channel) and P2Y (G protein-coupled) receptors. In this article we consider the dual role of ATP in regulation of vascular tone, released as a cotransmitter from sympathetic nerves or released in the vascular lumen in response to changes in blood flow and hypoxia. Further, purinergic long-term trophic and inflammatory signalling is described in cell proliferation, differentiation, migration and death in angiogenesis, vascular remodelling, restenosis and atherosclerosis. The effects on haemostasis and cardiac regulation is reviewed. The involvement of ATP in vascular diseases such as thrombosis, hypertension and diabetes will also be discussed, as well as various heart conditions. The purinergic system may be of similar importance as the sympathetic and renin-angiotensin-aldosterone systems in cardiovascular regulation and pathophysiology. The extracellular nucleotides and their cardiovascular P2 receptors are now entering the phase of clinical development.
Collapse
Affiliation(s)
- David Erlinge
- Department of Cardiology, Lund University Hospital, 22185, Lund, Sweden,
| | | |
Collapse
|
205
|
Characterization of a new peptide agonist of the protease-activated receptor-1. Biochem Pharmacol 2007; 75:438-47. [PMID: 17950254 DOI: 10.1016/j.bcp.2007.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 08/27/2007] [Accepted: 09/04/2007] [Indexed: 01/27/2023]
Abstract
A new peptide (TFRRRLSRATR), derived from the c-terminal of human platelet P2Y(1) receptor, was synthesized and its biological function was evaluated. This peptide activated platelets in a concentration-dependent manner, causing shape change, aggregation, secretion and calcium mobilization. Of the several receptor antagonists tested, only BMS200261, a protease activated receptor 1 (PAR-1) specific antagonist, totally abolished the peptide-induced platelet aggregation, secretion and calcium mobilization. The TFRRR-peptide-pretreated washed platelets failed to aggregate in response to SFLLRN (10 microM) but not to AYPGKF (500 microM). In addition, in mouse platelets, peptide concentrations up to 600 microM failed to cause platelet activation, indicating that the TFRRR-peptide activated platelets through the PAR-1 receptor, rather than through the PAR-4 receptor. The shape change induced by 10 microM peptide was totally abolished by Y-27632, an inhibitor of p160(ROCK) which is a downstream mediator of G12/13 pathways. The TFRRR-peptide, YFLLRNP, and the physiological agonist thrombin selectively activated G12/13 pathways at low concentrations and began to activate both Gq and G12/13 pathways with increasing concentrations. Similar to SFLLRN, the TFRRR-peptide caused phosphorylation of Akt and Erk in a P2Y(12) receptor-dependent manner, and p-38 MAP kinase activation in a P2Y(12)-independent manner. The effects of this peptide are elicited by the first six amino acids (TFRRRL) whereas the remaining peptide (LSRATR), TFERRN, or TFEERN had no effects on platelets. We conclude that TFRRRL activates human platelets through PAR-1 receptors.
Collapse
|
206
|
Kim S, Garcia A, Jackson SP, Kunapuli SP. Insulin-like growth factor-1 regulates platelet activation through PI3-Kalpha isoform. Blood 2007; 110:4206-13. [PMID: 17827385 PMCID: PMC2234779 DOI: 10.1182/blood-2007-03-080804] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Platelets release insulin-like growth factor-1 (IGF-1) from alpha granules upon activation. We have investigated the regulation of IGF-1 in G(i)-dependent pathways leading to Akt activation and the role of IGF-1 in platelet activation. IGF-1 alone failed to induce platelet aggregation, but IGF-1 potentiated 2-MeSADP-induced platelet aggregation in a concentration-dependent manner. IGF-1 triggered platelet aggregation in combination with selective P2Y(1) receptor activation. IGF-1 also caused platelet aggregation without shape change when combined with selective G(z) stimulation by epinephrine, suggesting the role of IGF-1 in platelet aggregation by supplementing G(i) pathways. The potentiating effect of IGF-1 was not affected by intracellular calcium chelation. Importantly, IGF-1 was unable to potentiate platelet aggregation by the phosphatidylinositol 3-kinase (PI3-K) inhibitor wortmannin, suggesting a critical regulation by PI3-K. Moreover, the potentiating effect of IGF-1 was abolished by the presence of PI3-K p110alpha inhibitor PIK-75. Stimulation of platelets with IGF-1 resulted in phosphorylation of Akt, a downstream effector of PI3-K, which was completely inhibited by wortmannin. IGF-1-induced Akt phosphorylation was abolished by PIK-75 suggesting the contribution of PI3-K p110alpha for activation of Akt by IGF-1. These results demonstrate that IGF-1 plays a role in potentiating platelet aggregation by complementing G(i)- but not G(q)-signaling pathways via PI3-K p110alpha.
Collapse
Affiliation(s)
- Soochong Kim
- Department of Physiology and Pharmacology and the Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | |
Collapse
|
207
|
Sagdilek E, Buyukcoskun NI, Ozluk K. Evaluation of platelet function and lack of response to epinephrine in pregnant women. Int J Lab Hematol 2007; 29:302-9. [PMID: 17617081 DOI: 10.1111/j.1365-2257.2006.00844.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Previous studies in healthy subjects have demonstrated a lack of response of platelets to epinephrine at a rate of 16-40% on an aggregometer. An association between the increased procoagulant factors during pregnancy and venous thromboembolism is known, and it has also been shown that prolactin levels increase platelet aggregation. We evaluated whether platelet functions in pregnant women and also assessed the lack of response to epinephrine during this period. We compared 27 healthy and volunteering pregnant women with 26 similar control subjects. Platelet functions were assessed with an aggregometer and a Platelet Function Analyzer (PFA-100). Less than 40% response to epinephrine on the aggregometer was defined as an impaired epinephrine response. The aggregation response of epinephrine was normal in 25 of the 27 pregnant women, while two of them showed a late-rising response. Eight of the 26 subject control group (30.8%) showed an impaired response to epinephrine. When we compared the 25 pregnant and 18 control subjects with normal aggregation responses, the maximum aggregation responses to ADP and epinephrine, and the Col/Epi and Col/ADP cartridge closure time values were significantly lower in pregnant women. There were no difference between second and third trimesters as regards platelet function parameters. The fact that no impaired response to epinephrine was detected in pregnant women while a 30% rate was observed in non-pregnant women indicates that the platelet malfunction caused by a disorder in the Gi protein and intracellular mechanisms is bypassed during pregnancy thanks to some physiological changes.
Collapse
Affiliation(s)
- E Sagdilek
- Department of Physiology, Faculty of Medicine, Uludag University, Bursa, Turkey.
| | | | | |
Collapse
|
208
|
Garcia A, Shankar H, Murugappan S, Kim S, Kunapuli S. Regulation and functional consequences of ADP receptor-mediated ERK2 activation in platelets. Biochem J 2007; 404:299-308. [PMID: 17298299 PMCID: PMC1868805 DOI: 10.1042/bj20061584] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have previously shown that ADP-induced thromboxane generation in platelets requires signalling events from the G(q)-coupled P2Y1 receptor (platelet ADP receptor coupled to stimulation of phospholipase C) and the G(i)-coupled P2Y12 receptor (platelet ADP receptor coupled to inhibition of adenylate cyclase) in addition to outside-in signalling. While it is also known that extracellular calcium negatively regulates ADP-induced thromboxane A2 generation, the underlying mechanism remains unclear. In the present study we sought to elucidate the signalling mechanisms and regulation by extracellular calcium of ADP-induced thromboxane A2 generation in platelets. ERK (extracllular-signal-regulated kinase) 2 activation occurred when outside-in signalling was blocked, indicating that it is a downstream event from the P2Y receptors. However, blockade of either P2Y1 or the P2Y12 receptors with corresponding antagonists completely abolished ERK phosphorylation, indicating that both P2Y receptors are required for ADP-induced ERK activation. Inhibitors of Src family kinases or the ERK upstream kinase MEK [MAPK (mitogen-activated protein kinase)/ERK kinase] abrogated ADP-induced ERK phosphorylation and thromboxane A2 generation. Finally ADP- or G(i)+G(z)-induced ERK phosphorylation was blocked in the presence of extracellular calcium. The present studies show that ERK2 is activated downstream of P2Y receptors through a complex mechanism involving Src kinases and this plays an important role in ADP-induced thromboxane A2 generation. We also conclude that extracellular calcium blocks ADP-induced thromboxane A2 generation through the inhibition of ERK activation.
Collapse
Affiliation(s)
- Analia Garcia
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Haripriya Shankar
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Swaminathan Murugappan
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Soochong Kim
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Satya P. Kunapuli
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- ‡Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
209
|
Coppinger JA, O'Connor R, Wynne K, Flanagan M, Sullivan M, Maguire PB, Fitzgerald DJ, Cagney G. Moderation of the platelet releasate response by aspirin. Blood 2007; 109:4786-92. [PMID: 17303692 DOI: 10.1182/blood-2006-07-038539] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Modulation of the proteins released during activation is one mechanism whereby aspirin may influence platelet-mediated human disease. We investigated the effect of aspirin on the platelet releasate using mass spectrometry and found that different agonists evoked different releasate profiles, with aspirin having a general moderating effect on the amount of protein released regardless of the agonist. These observations were confirmed for several cytokines using an antibody array approach.
Collapse
Affiliation(s)
- Judith A Coppinger
- Department of Cell Biology, Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
210
|
Abstract
Platelets play a central role in hemostasis, but also in atherothrombosis, as they rapidly adhere to tissue and to one another as a response to any vascular injury. This process involves a large number of surface receptors, signaling pathways, and enzymatic cascades as well as their complex interplay. Although in vitro experiments proved successful in both identifying new receptors and pathways and developing potent and selective antithrombotic drugs, in vitro research cannot mimic the myriad hemodynamic and spatiotemporal cellular and molecular interactions that occur during the generation and propagation of thrombi in vivo. Animal models, and, with the availability of genetically modified mouse strains and of modern intravital imaging techniques, mouse models in particular, have opened new ways to identify both individual roles and the interplay of platelet proteins in complex in vivo settings. In vivo models revealed the important role of, eg, Gas6 or blood coagulation factor XII in thrombus formation, and results obtained in in vivo models raised the interesting possibility that (physiologic) hemostasis and (pathologic) thrombosis might represent 2 mechanistically different processes. This review summarizes in vivo findings that contributed significantly to our understanding of hemostatic and thrombotic processes and which may help to guide future research.
Collapse
Affiliation(s)
- Ulrich J H Sachs
- Rudolf Virchow Center, Deutsche Forschungsgemeinschaft Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | | |
Collapse
|
211
|
Strehl A, Munnix ICA, Kuijpers MJE, van der Meijden PEJ, Cosemans JMEM, Feijge MAH, Nieswandt B, Heemskerk JWM. Dual Role of Platelet Protein Kinase C in Thrombus Formation. J Biol Chem 2007; 282:7046-55. [PMID: 17210570 DOI: 10.1074/jbc.m611367200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Protein kinase C (PKC) isoforms regulate many platelet responses in a still incompletely understood manner. Here we investigated the roles of PKC in the platelet reactions implicated in thrombus formation as follows: secretion aggregate formation and coagulation-stimulating activity, using inhibitors with proven activity in plasma. In human and mouse platelets, PKC regulated aggregation by mediating secretion and contributing to alphaIIbbeta3 activation. Strikingly, PKC suppressed Ca(2+) signal generation and Ca(2+)-dependent exposure of procoagulant phosphatidylserine. Furthermore, under coagulant conditions, PKC suppressed the thrombin-generating capacity of platelets. In flowing human and mouse blood, PKC contributed to platelet adhesion and controlled secretion-dependent thrombus formation, whereas it down-regulated Ca(2+) signaling and procoagulant activity. In murine platelets lacking G(q)alpha, where secretion reactions were reduced in comparison with wild type mice, PKC still positively regulated platelet aggregation and down-regulated procoagulant activity. We conclude that platelet PKC isoforms have a dual controlling role in thrombus formation as follows: (i) by mediating secretion and integrin activation required for platelet aggregation under flow, and (ii) by suppressing Ca(2+)-dependent phosphatidylserine exposure, and consequently thrombin generation and coagulation. This platelet signaling protein is the first one identified to balance the pro-aggregatory and procoagulant functions of thrombi.
Collapse
Affiliation(s)
- Amrei Strehl
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, University of Maastricht, 6200 MD Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
212
|
Abstract
Because of their ability to become rapidly activated at places of vascular injury, platelets are important players in primary hemostasis as well as in arterial thrombosis. In addition, they are also involved in chronic pathological processes including the atherosclerotic remodeling of the vascular system. Although primary adhesion of platelets to the vessel wall is largely independent of G protein-mediated signaling, the subsequent recruitment of additional platelets into a growing platelet thrombus requires mediators such as ADP, thromboxane A(2), or thrombin, which act through G protein-coupled receptors. Platelet activation via G protein-coupled receptors involves 3 major G protein-mediated signaling pathways that are initiated by the activation of the G proteins G(q), G(13), and G(i). This review summarizes recent progress in understanding the mechanisms underlying platelet activation and thrombus extension via G protein-mediated signaling pathways.
Collapse
Affiliation(s)
- Stefan Offermanns
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
213
|
|
214
|
|
215
|
Eriksson AC, Whiss PA, Nilsson UK. Adhesion of human platelets to albumin is synergistically increased by lysophosphatidic acid and adrenaline in a donor-dependent fashion. Blood Coagul Fibrinolysis 2006; 17:359-68. [PMID: 16788312 DOI: 10.1097/01.mbc.0000233366.18605.b2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lysophosphatidic acid (LPA) and adrenaline are weak platelet activators considered important for thrombus formation, and were previously shown to synergistically increase platelet aggregation. Here we investigate synergistic activation by LPA and adrenaline when measuring platelet adhesion. Platelet-rich plasma from healthy blood donors together with adrenaline and/or LPA were added to protein-coated microplates. Platelets were allowed to adhere and the amount of adhesion detected enzymatically. The LPA and adrenaline combination induced a synergistic increase of platelet adhesion to a normally non-adhesive albumin surface. The degree of synergy varied markedly between individuals; these variations could not be explained by age, gender, blood type or different amounts of platelets, oxidized low-density lipoprotein, insulin or glucose in plasma. There was a trend indicating increased synergistic effect for platelets sensitive to adrenaline stimulation. The synergistic effect was blocked by the alpha2-adrenoceptor antagonist yohimbine and inhibited by the ADP scavenger system creatine phosphate/creatine phosphokinase and antibodies against alphaIIbbeta3. Furthermore, platelets adhering to albumin after adrenaline and LPA treatment expressed P-selectin. In conclusion, LPA and adrenaline act synergistically to increase alphaIIbbeta3-mediated platelet adhesion to albumin, dependent on alpha2-adrenoceptor signalling and platelet secretion. We also confirm that synergistic platelet activation achieved with LPA and adrenaline is highly donor dependent.
Collapse
Affiliation(s)
- Andreas C Eriksson
- Division of Pharmacology, Department of Medicine and Care, Faculty of Health Sciences, Linköping University, Sweden.
| | | | | |
Collapse
|
216
|
Minuz P, Fumagalli L, Gaino S, Tommasoli R, Degan M, Cavallini C, Lecchi A, Cattaneo M, Lechi Santonastaso C, Berton G. Rapid stimulation of tyrosine phosphorylation signals downstream of G-protein-coupled receptors for thromboxane A2 in human platelets. Biochem J 2006; 400:127-34. [PMID: 16859489 PMCID: PMC1635449 DOI: 10.1042/bj20061015] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Signals ensuing from trimeric G-protein-coupled receptors synergize to induce platelet activation. At low doses, the thromboxane A2 analogue U46619 does not activate integrin alphaIIbbeta3 or trigger platelet aggregation, but it induces shape changes. In the present study, we addressed whether low doses of U46619 trigger tyrosine phosphorylation independently of integrin alphaIIbbeta3 activation and ADP secretion, and synergize with adrenaline (epinephrine) to induce aggregation in acetylsalicylic acid (aspirin)-treated platelets. Low doses of U46619 triggered tyrosine phosphorylation of different proteins, including FAK (focal adhesion kinase), Src and Syk, independently of signals ensuing from integrin alphaIIbbeta3 or ADP receptors engaged by secreted ADP. The G(12/13)-mediated Rho/Rho-kinase pathway was also increased by low doses of U46619; however, this pathway was not upstream of tyrosine phosphorylation, because this occurred in the presence of the Rho-kinase inhibitor Y-27632. Although low doses of U46619 or adrenaline alone were unable to trigger platelet aggregation and integrin alphaIIbbeta3 activation, the combination of the two stimuli effectively induced these responses. PP2, a tyrosine kinase inhibitor, and Y-27632 inhibited platelet activation induced by low doses of U46619 plus adrenaline and, when used in combination, totally suppressed this platelet response. In addition, the two inhibitors selectively blocked tyrosine kinases and the Rho/Rho-kinase pathway respectively. These findings suggest that both tyrosine phosphorylation and the Rho/Rho-kinase pathway are required to activate platelet aggregation via G(12/13) plus G(z) signalling.
Collapse
Affiliation(s)
- Pietro Minuz
- *Department of Biomedical and Surgical Sciences, University of Verona, Verona, Italy
| | - Laura Fumagalli
- †Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy
| | - Stefania Gaino
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Rosa M. Tommasoli
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Maurizio Degan
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Chiara Cavallini
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Anna Lecchi
- §Angelo Bianchi Bonomi Haemophila and Thrombosis Centre, IRCCS Maggiore Hospital, University of Milan, Milan, Italy
| | - Marco Cattaneo
- ∥Unit of Haematology and Thrombosis, San Paolo Hospital, DMCO-University of Milan, Milan, Italy
| | | | - Giorgio Berton
- †Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy
- To whom correspondence should be addressed (email )
| |
Collapse
|
217
|
Kahner BN, Shankar H, Murugappan S, Prasad GL, Kunapuli SP. Nucleotide receptor signaling in platelets. J Thromb Haemost 2006; 4:2317-26. [PMID: 17059469 DOI: 10.1111/j.1538-7836.2006.02192.x] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
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.
Collapse
Affiliation(s)
- B N Kahner
- The Cell Signaling Group, Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
218
|
Nguyen T, Frishman WH, Nawarskas J, Lerner RG. Variability of response to clopidogrel: possible mechanisms and clinical implications. Cardiol Rev 2006; 14:136-42. [PMID: 16628022 DOI: 10.1097/01.crd.0000188033.11188.8e] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Clopidogrel has been shown to inhibit adenosine diphosphate-induced platelet aggregation and has been demonstrated to be effective in reducing the risk of arterial thrombotic events in several large clinical studies. However, the clinical benefit could be attenuated by the variability of response to the antiplatelet effects of clopidogrel in as many as 30% of patients. Multiple mechanisms likely contribute to clopidogrel variability of response, including inappropriate dosing or underdosing of clopidogrel, drug-drug interactions, and genetic polymorphisms. The best laboratory procedure to screen for possible clopidogrel variability of response remains to be determined.
Collapse
Affiliation(s)
- Theresa Nguyen
- Department of Medicine, Virginia Mason Medical Center, Seattle, Washington, USA
| | | | | | | |
Collapse
|
219
|
Nylander S, Mattsson C, Lindahl TL. Characterisation of species differences in the platelet ADP and thrombin response. Thromb Res 2006; 117:543-9. [PMID: 15921723 DOI: 10.1016/j.thromres.2005.04.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2004] [Revised: 04/12/2005] [Accepted: 04/22/2005] [Indexed: 11/19/2022]
Abstract
INTRODUCTION A number of animal models are used to study platelet-dependent diseases. In the present investigation, we have used a simple flow cytometry assay to evaluate platelet function in man, rat, mouse, guinea pig and dog. MATERIALS AND METHODS Platelet activation was evaluated in diluted whole blood by measuring fibrinogen binding to activated platelets using a polyclonal anti-human fibrinogen antibody that cross-reacts with fibrinogen from all species tested. The assay was used to evaluate platelet function with respect to ADP and thrombin sensitivity. The relative importance of the two platelet ADP receptors and total ADP in the thrombin response was also studied by using receptor-specific antagonists and apyrase, respectively. RESULTS Mouse platelets were most sensitive to both agonists. Unlike in man and dog the maximal response to ADP was greater than to thrombin in mouse, rat and guinea pig. P2Y(12) blockade was in all species equally effective as ADP removal in inhibiting thrombin-induced platelet activation whereas P2Y(1) blockade was almost ineffective. CONCLUSION The present study describes a simple platelet function test that can be used to evaluate platelet function in man, rat, mouse, guinea pig and dog. Platelets from the tested species differed in their sensitivity to ADP and thrombin. In contrast to human and canine platelets, mouse, rat and guinea pig platelets displayed a stronger maximal response to ADP than to thrombin. In terms of the relative contribution of P2Y(1) and P2Y(12) in the thrombin response, the P2Y(12) receptor was the key receptor in all species and its blockade gave equal effect as total removal of ADP.
Collapse
Affiliation(s)
- Sven Nylander
- Department of Molecular Pharmacology, Preclinical R and D, AstraZeneca R and D, Mölndal, 431 83 Mölndal, Sweden.
| | | | | |
Collapse
|
220
|
Abbracchio MP, Burnstock G, Boeynaems JM, Barnard EA, Boyer JL, Kennedy C, Knight GE, Fumagalli M, Gachet C, Jacobson KA, Weisman GA. International Union of Pharmacology LVIII: update on the P2Y G protein-coupled nucleotide receptors: from molecular mechanisms and pathophysiology to therapy. Pharmacol Rev 2006; 58:281-341. [PMID: 16968944 PMCID: PMC3471216 DOI: 10.1124/pr.58.3.3] [Citation(s) in RCA: 999] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
There have been many advances in our knowledge about different aspects of P2Y receptor signaling since the last review published by our International Union of Pharmacology subcommittee. More receptor subtypes have been cloned and characterized and most orphan receptors de-orphanized, so that it is now possible to provide a basis for a future subdivision of P2Y receptor subtypes. More is known about the functional elements of the P2Y receptor molecules and the signaling pathways involved, including interactions with ion channels. There have been substantial developments in the design of selective agonists and antagonists to some of the P2Y receptor subtypes. There are new findings about the mechanisms underlying nucleotide release and ectoenzymatic nucleotide breakdown. Interactions between P2Y receptors and receptors to other signaling molecules have been explored as well as P2Y-mediated control of gene transcription. The distribution and roles of P2Y receptor subtypes in many different cell types are better understood and P2Y receptor-related compounds are being explored for therapeutic purposes. These and other advances are discussed in the present review.
Collapse
Affiliation(s)
- Maria P Abbracchio
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Hasan KN, Shoji M, Tsutaya S, Kudo R, Matsuda E, Saito J, Kimura T, Yasujima M. Study of V1a vasopressin receptor gene single nucleotide polymorphisms in platelet vasopressin responsiveness. J Clin Lab Anal 2006; 20:87-92. [PMID: 16721832 PMCID: PMC6807323 DOI: 10.1002/jcla.20106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
There is a significant heterogeneity among individuals in terms of platelet aggregation response to arginine vasopressin (AVP). The aim of this study was to evaluate whether four single nucleotide polymorphisms (SNPs) in the promoter region of vasopressin V1a receptor gene (V1aR) could be used as genetic markers for divergent platelet aggregation response to AVP. Seventeen of 33 subjects showed more than 60% of maximum platelet aggregation and were classified as responders. Sixteen were classified as nonresponders because they had less than 30% aggregation. In a preliminary study, V1aR gene sequences were determined in two responders and two nonresponders. We found four SNPs in the promoter region of the V1aR gene: -6951G/A, -4112A/T, -3860T/C, and -242C/T. In all 33 subjects the genotypes of four SNPs were determined using either polymerase chain reaction (PCR) with allele-specific primers or PCR followed by restriction-fragment length polymorphism (RFLP). There were no differences in the AVP-induced aggregation between the subjects with and without variant alleles of each four SNPs. The genotype frequencies of four SNPs of V1aR were almost identical between AVP responders and nonresponders. These results suggest that the four SNPs in the promoter region of the V1aR gene may not be useful as genetic markers for platelet aggregation heterogeneity.
Collapse
Affiliation(s)
- Kazi N. Hasan
- Department of Laboratory Medicine, Hirosaki University School of Medicine, Hirosaki University, Hirosaki, Japan
| | - Masaru Shoji
- Department of Laboratory Medicine, Hirosaki University School of Medicine, Hirosaki University, Hirosaki, Japan
| | - Shoji Tsutaya
- Department of Clinical Laboratory, Hirosaki University Hospital, Hirosaki, Japan
| | - Ryoko Kudo
- Department of Clinical Laboratory, Hirosaki University Hospital, Hirosaki, Japan
| | - Eriko Matsuda
- Department of Clinical Laboratory, Hirosaki University Hospital, Hirosaki, Japan
| | - Junko Saito
- Department of Clinical Laboratory, Hirosaki University Hospital, Hirosaki, Japan
| | | | - Minoru Yasujima
- Department of Laboratory Medicine, Hirosaki University School of Medicine, Hirosaki University, Hirosaki, Japan
| |
Collapse
|
222
|
Shankar H, Kahner BN, Prabhakar J, Lakhani P, Kim S, Kunapuli SP. G-protein-gated inwardly rectifying potassium channels regulate ADP-induced cPLA2 activity in platelets through Src family kinases. Blood 2006; 108:3027-34. [PMID: 16857990 PMCID: PMC1895524 DOI: 10.1182/blood-2006-03-010330] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ADP-induced TXA2 generation requires the costimulation of P2Y1, P2Y12, and the GPIIb/IIIa receptors. Signaling events downstream of the P2Y receptors that contribute to ADP-induced TXA2 generation have not been clearly delineated. In this study, we have investigated the role of G-protein-gated inwardly rectifying potassium channels (GIRKs), a recently identified functional effector for the P2Y12 receptor, in the regulation of ADP-induced TXA2 generation. At 10-microM concentrations, the 2 structurally distinct GIRK channel blockers, SCH23390 and U50488H, caused complete inhibition of ADP-induced cPLA2 phosphorylation and TXA2 generation, without affecting the conversion of AA to TXA2 or ADP-induced primary platelet aggregation in aspirin-treated platelets. In addition, Src family kinase selective inhibitors abolished 2MeSADP-mediated cPLA2 phosphorylation and TXA2 generation. Furthermore, these GIRK channel blockers completely blocked Gi-mediated Src kinase activation, suggesting that GIRK channels are upstream of Src family tyrosine kinase activation. In weaver mouse platelets, which have dysfunctional GIRK2 subunits, ADP-induced TXA2 generation was impaired. However, we did not observe any defect in 2MeSADP-induced platelet functional responses in GIRK2-null mouse platelets, suggesting that functional channels composed of other GIRK subunits contribute to ADP-induced TXA2 generation, via the regulation of the Src and cPLA2 activity.
Collapse
Affiliation(s)
- Haripriya Shankar
- Department of Physiology, Sol Sherry Thrombosis Research Center, Temple University, Rm 224OMS, 3420 N Broad St, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
223
|
Savi P, Zachayus JL, Delesque-Touchard N, Labouret C, Hervé C, Uzabiaga MF, Pereillo JM, Culouscou JM, Bono F, Ferrara P, Herbert JM. The active metabolite of Clopidogrel disrupts P2Y12 receptor oligomers and partitions them out of lipid rafts. Proc Natl Acad Sci U S A 2006; 103:11069-74. [PMID: 16835302 PMCID: PMC1635153 DOI: 10.1073/pnas.0510446103] [Citation(s) in RCA: 235] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
P2Y12, a G protein-coupled receptor that plays a central role in platelet activation has been recently identified as the receptor targeted by the antithrombotic drug, clopidogrel. In this study, we further deciphered the mechanism of action of clopidogrel and of its active metabolite (Act-Met) on P2Y12 receptors. Using biochemical approaches, we demonstrated the existence of homooligomeric complexes of P2Y12 receptors at the surface of mammalian cells and in freshly isolated platelets. In vitro treatment with Act-Met or in vivo oral administration to rats with clopidogrel induced the breakdown of these oligomers into dimeric and monomeric entities in P2Y12 expressing HEK293 and platelets respectively. In addition, we showed the predominant association of P2Y12 oligomers to cell membrane lipid rafts and the partitioning of P2Y12 out of rafts in response to clopidogrel and Act-Met. The raft-associated P2Y12 oligomers represented the functional form of the receptor, as demonstrated by binding and signal transduction studies. Finally, using a series of receptors individually mutated at each cysteine residue and a chimeric P2Y12/P2Y13 receptor, we pointed out the involvement of cysteine 97 within the first extracellular loop of P2Y12 in the mechanism of action of Act-Met.
Collapse
Affiliation(s)
- Pierre Savi
- Department of Thrombosis and Angiogenesis, Sanofi-Aventis Recherche, 195 Route d'Espagne, 31036 Toulouse, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Rossi R, Giustarini D, Dalle-Donne I, Milzani A. Protein S-glutathionylation and platelet anti-aggregating activity of disulfiram. Biochem Pharmacol 2006; 72:608-15. [PMID: 16815310 DOI: 10.1016/j.bcp.2006.05.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 05/26/2006] [Accepted: 05/26/2006] [Indexed: 02/02/2023]
Abstract
Blood platelets are central to haemostasis, and reactions in platelets involving sulfhydryl groups play important roles in platelet function. Reduced glutathione (GSH) plays an important role in platelet aggregation and glutathione-depleting chemicals inhibit platelet aggregation. The lipophilic drug disulfiram, because of its affinity for sulfhydryl groups, is a highly thiol-reacting agent. As a consequence, GSH and sulfhydryl groups of protein cysteines in human platelets, in analogy to other components of human blood, are a potential target of disulfiram. In the present study, we have shown that exposure of human platelets to disulfiram causes the depletion of platelet GSH and augmentation of mixed disulfides between GSH and protein sulfhydryl groups to form protein-glutathione mixed disulfides (S-glutathionylated proteins). The depletion of platelet GSH and the increase in S-glutathionylated proteins occurred at concentrations of disulfiram that inhibited platelet aggregation, suggesting that protein S-glutathionylation is involved in the inhibition of platelet aggregation caused by disulfiram.
Collapse
Affiliation(s)
- Ranieri Rossi
- Department of Neuroscience, University of Siena, I-53100 Siena, Italy
| | | | | | | |
Collapse
|
225
|
Abstract
The main role of blood platelets is to ensure primary hemostasis, which is the maintenance of vessel integrity and cessation of bleeding upon injury. While playing a major part in acute arterial thrombosis, platelets are also involved in inflammation, atherosclerosis, and angiogenesis. ADP and ATP play a crucial role in platelet activation, and their receptors are potential targets for antithrombotic drugs. The ATP-gated cation channel P2X(1) and the two G protein-coupled ADP receptors, P2Y(1) and P2Y(12), selectively contribute to platelet aggregation and formation of a thrombus. Owing to its central role in the growth and stabilization of a thrombus, the P2Y(12) receptor is an established target of antithrombotic drugs such as clopidogrel. Studies in P2Y(1) and P2X(1) knockout mice and selective P2Y(1) and P2X(1) antagonists have shown that these receptors are also attractive targets for new antithrombotic compounds. The potential role of platelet P(2) receptors in the involvement of platelets in inflammatory processes is also discussed.
Collapse
Affiliation(s)
- Christian Gachet
- Institut National de la Santé et de la Recherche Médicale, Unité 311, Etablissement Français du Sang-Alsace, Strasbourg 67065, France.
| |
Collapse
|
226
|
Parvizpur A, Ahmadiani A, Kamalinejad M. Probable role of spinal purinoceptors in the analgesic effect of Trigonella foenum (TFG) leaves extract. JOURNAL OF ETHNOPHARMACOLOGY 2006; 104:108-12. [PMID: 16298092 DOI: 10.1016/j.jep.2005.08.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2004] [Revised: 08/05/2005] [Accepted: 08/26/2005] [Indexed: 05/05/2023]
Abstract
In our previous work, we demonstrated that Trigonella foenum (TFG) leaves extract can exert analgesic effects in both formalin (F.T.) and tail flick (T.F.) tests. Spinal serotonergic system, but not endogenous opioid system, was involved in TFG induced analgesia (in the second phase of formalin test). Some reports concern the similarity between NSAIDs and TFG extract in many pharmacological effects or the interaction between NSAIDs and purinergic system; so the present study was designed to investigate the relationship between TFG extract and purinergic system or the inhibition of cyclo-oxygenase (COX). We examined the effect of TFG extract on: (1) the response of rabbit platelets to ADP induced aggregation, (2) the contraction of mouse vas deferens induced by alpha,beta-Me-ATP (a P(2) receptor agonist; this receptor mediates the rapid phase of ADP- and ATP-evoked influx of Ca(2+) through a non-specific cation channel in platelets), (3) alpha,beta-Me-ATP induced hyperalgesia in tail flick test in male rats and (4) the specific inhibition of COX-1 and COX-2. Our results showed that TFG extract (0.5, 1, 1.5, 3 mg/ml) inhibited ADP (10(-5) mol) induced platelet aggregation (IC(50)=1.28 mg/ml). alpha,beta-Me-ATP (30 microM) induced isometric contraction in vas deferens while suramin (a P(2) receptor antagonist, 50, 150, 300 microM) or TFG extract (0.5, 1, 2, 3 mg/ml) inhibited this effect significantly (IC(50) were 91.07 microM and 1.57 mg/ml, respectively). Moreover, alpha,beta-Me-ATP (3 microg/rat, i.t.) induced hyperalgesia in tail flick test, but it was prevented by co-injection of alpha,beta-Me-ATP with suramin (120 microg/rat, i.t.) or TFG extract (1mg/rat, i.t.). Effective concentrations of TFG extract in the above mentioned experiments did not inhibit COX enzymes in EIA tests. In conclusion, these results indicate that the blocking of spinal purinoceptors may contribute in the analgesic effect of TFG leaves extract.
Collapse
Affiliation(s)
- Aliresa Parvizpur
- Neuroscience Research Center, Department of Pharmacology, School of Medicine, Shaheed Beheshti University of Medical Sciences, P.O. Box 19835-355, Tehran, Iran
| | | | | |
Collapse
|
227
|
Abstract
Hemostasis is a normal process preventing the sequelae of uncontrolled hemorrhage. In certain settings, these same processes cause adverse clinical events due to thrombotic occlusion of a vessel. The majority of unstable coronary syndromes result from disruption of an atherosclerotic plaque, leading to the exposure of subintimal contents, which triggers coagulation and the formation of a platelet-rich thrombus. The central role of platelet activation in the events that lead to vessel occlusion is well known. However, this process is complex and influenced by a myriad of cellular and plasma-derived mediators that regulate the balance between occlusive and nonocclusive thrombosis.
Collapse
Affiliation(s)
- Jane E Freedman
- Boston University School of Medicine, Whitaker Cardiovascular Institute, Boston, MA 02118, USA.
| |
Collapse
|
228
|
Gachet C, Léon C, Hechler B. The platelet P2 receptors in arterial thrombosis. Blood Cells Mol Dis 2006; 36:223-7. [PMID: 16466948 DOI: 10.1016/j.bcmd.2005.12.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 12/07/2005] [Indexed: 11/24/2022]
Abstract
ADP and ATP play a crucial role in hemostasis and thrombosis and their receptors are potential targets for antithrombotic drugs. The ATP-gated channel P2X1 and the two G protein-coupled P2Y1 and P2Y12 ADP receptors selectively contribute to platelet aggregation. Due to its central role in the formation and stabilization of a thrombus, the P2Y12 receptor is a well established target of antithrombotic drugs like clopidogrel which has proved efficacious in many clinical trials and experimental models of thrombosis. Competitive P2Y12 antagonists have also been shown to be effective in experimental thrombosis as well as in several clinical trials. Studies in P2Y1 and P2X1 knock-out mice and experimental thrombosis models using selective P2Y1 and P2X1 antagonists have shown that, depending on the conditions, these receptors could also be potential targets for new antithrombotic drugs. Since both P2X1 and P2Y1 receptor inhibition result in milder prolongation of the bleeding time as compared to P2Y12 inhibition, the idea is put forward that combination of P2 receptor antagonists could improve efficacy with diminished hemorrhagic risk. However, further studies are required to validate such a point of view.
Collapse
Affiliation(s)
- Christian Gachet
- Institut National de la Santé et de la Recherche Médicale, Unité 311, Etablissement Français du Sang-Alsace, Strasbourg, France.
| | | | | |
Collapse
|
229
|
Ding Z, Kim S, Kunapuli SP. Identification of a potent inverse agonist at a constitutively active mutant of human P2Y12 receptor. Mol Pharmacol 2006; 69:338-45. [PMID: 16234484 DOI: 10.1124/mol.105.014654] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human platelets express two P2Y receptors: G(q)-coupled P2Y(1), and G(i)-coupled P2Y(12). Both P2Y(1) and P2Y(12) are ADP receptors on human platelets and are essential for ADP-induced platelet aggregation that plays pivotal roles in thrombosis and hemostasis. Numerous constitutively active G protein-coupled receptors have been described in natural or recombinant systems, but in the P2Y receptors, to date, no constitutive activity has been reported. In our effort to identify G protein coupling domains of the human platelet ADP receptor, we constructed a chimeric hemagglutinin-tagged human P2Y(12) receptor with its C terminus replaced by the corresponding part of human P2Y(1) receptor and stably expressed it in Chinese hamster ovary-K1 cells. It is interesting that the chimeric P2Y(12) mutant exhibited a high level of constitutive activity, as evidenced by decreased cAMP levels in the absence of agonists. The constitutive activation of the chimeric P2Y(12) mutant was dramatically inhibited by pertussis toxin, a G(i) inhibitor. The constitutively active P2Y(12) mutant retained normal responses to 2-methylthio-ADP, with an EC(50) of 0.15 +/- 0.04 nM. The constitutively active P2Y(12) mutant caused Akt phosphorylation that was abolished by the addition of pertussis toxin. Pharmacological evaluation of several P2Y(12) antagonists revealed (E)-N-[1-[7-(hexylamino)-5-(propylthio)-3H-1,2,3-triazolo-[4,5-d]-pyrimidin-3-yl]-1,5,6-trideoxy-beta-d-ribo-hept-5-enofuranuronoyl]-l-aspartic acid (AR-C78511) as a potent P2Y(12) inverse agonist and 5'-adenylic acid, N-[2-(methylthio)ethyl]-2-[(3,3,3-trifluoropropyl)thio]-, monoanhydride with (dichloromethylene)bis[phosphonic acid] (AR-C69931MX) as a neutral antagonist. In conclusion, this is the first report of a cell line stably expressing a constitutively active mutant of human platelet P2Y(12) receptor and the identification of potent inverse agonist.
Collapse
Affiliation(s)
- Zhongren Ding
- Department of Physiology, Temple University School of Medicine, 3420 N. Broad Street, Philadelphia, PA 19140, USA
| | | | | |
Collapse
|
230
|
Coomber BL, Mitchell GB, Starr AE, Minhas K, Tamblyn A, Shewen PE, Gentry PA. Clopidogrel induced suppression of bovine platelet activation in vitro and a preliminary study of its effect on the development of Mannheimia haemolytica induced pneumonia. Vet J 2006; 171:126-34. [PMID: 16427590 DOI: 10.1016/j.tvjl.2004.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2004] [Indexed: 11/17/2022]
Abstract
We report here on the influence of the platelet antagonist clopidogrel (Plavix) on bovine platelet function. We first evaluated the capacity of clopidogrel to inhibit adenosine diphosphate (ADP)-stimulated platelet function in the bovine species, using an ex vivo approach with blood from treated animals. Platelets isolated from treated calves displayed rapid and consistent reduction in function (aggregation, thromboxane production) upon ADP, but not platelet activating factor (PAF), stimulation. We then examined the possibility that clopidogrel could influence Mannheimia haemolytica pneumonia pathobiology using an experimental challenge model. We were unable to detect significant differences between clopidogrel treated and untreated animals when challenged with intra-tracheal inoculation of M. haemolytica. There was a trend towards inhibition of platelet degranulation in the affected regions of lungs from clopidogrel treated calves, and pre-treated challenged animals had similar amounts of fibrin deposition and enhanced fibrous tissue formation in their lungs when compared with control counterparts.
Collapse
Affiliation(s)
- Brenda L Coomber
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ont., Canada N1G 2W1.
| | | | | | | | | | | | | |
Collapse
|
231
|
Freedman JE, Loscalzo J. Thrombosis. Vasc Med 2006. [DOI: 10.1016/b978-0-7216-0284-4.50014-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
232
|
Abstract
Heterotrimeric G proteins are key players in transmembrane signaling by coupling a huge variety of receptors to channel proteins, enzymes, and other effector molecules. Multiple subforms of G proteins together with receptors, effectors, and various regulatory proteins represent the components of a highly versatile signal transduction system. G protein-mediated signaling is employed by virtually all cells in the mammalian organism and is centrally involved in diverse physiological functions such as perception of sensory information, modulation of synaptic transmission, hormone release and actions, regulation of cell contraction and migration, or cell growth and differentiation. In this review, some of the functions of heterotrimeric G proteins in defined cells and tissues are described.
Collapse
Affiliation(s)
- Nina Wettschureck
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | | |
Collapse
|
233
|
Kim S, Jin J, Kunapuli SP. Relative contribution of G-protein-coupled pathways to protease-activated receptor-mediated Akt phosphorylation in platelets. Blood 2005; 107:947-54. [PMID: 16223779 DOI: 10.1182/blood-2005-07-3040] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protease-activated receptors (PARs) activate Gq and G(12/13) pathways, as well as Akt (protein kinase B [PKB/Akt]) in platelets. However, the relative contribution of different G-protein pathways to Akt phosphorylation has not been elucidated. We investigated the contribution of Gq and G(12/13) to Gi/Gz-mediated Akt phosphorylation downstream of PAR activation. Selective G(12/13) activation failed to cause Akt phosphorylation in human and Galpha q-deficient mouse platelets. However, supplementing Gi/Gz signaling to G(12/13) caused significant increase in Akt phosphorylation, confirming that G(12/13) potentiates Akt phosphorylation. Inhibition of PAR-mediated Akt phosphorylation in the presence of the Gq-selective inhibitor YM-254890 was restored to the normal extent achieved by PAR agonists if supplemented with Gi signaling, indicating that Gq does not have any direct effect on Akt phosphorylation. Selective G(12/13) activation resulted in Src kinase activation, and Akt phosphorylation induced by costimulation of G(12/13) and Gi/Gz was inhibited by a Src kinase inhibitor but not by a Rho kinase inhibitor. These data demonstrate that G(12/13), but not Gq, is essential for thrombin-induced Akt phosphorylation in platelets, whereas Gq indirectly contributes to Akt phosphorylation through Gi stimulation by secreted ADP. G(12/13) activation might mediate its potentiating effect through Src activation, and Src kinases play an important role in thrombin-mediated Akt phosphorylation.
Collapse
Affiliation(s)
- Soochong Kim
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | |
Collapse
|
234
|
Inoue O, Suzuki-Inoue K, McCarty OJT, Moroi M, Ruggeri ZM, Kunicki TJ, Ozaki Y, Watson SP. Laminin stimulates spreading of platelets through integrin alpha6beta1-dependent activation of GPVI. Blood 2005; 107:1405-12. [PMID: 16219796 PMCID: PMC1895394 DOI: 10.1182/blood-2005-06-2406] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix protein, laminin, supports platelet adhesion through binding to integrin alpha6beta1 In the present study, we demonstrate that human laminin, purified from placenta, also stimulates formation of filopodia and lamellipodia in human and mouse platelets through a pathway that is dependent on alpha6beta1 and the collagen receptor GPVI. The integrin alpha6beta1 is essential for adhesion to laminin, as demonstrated using an alpha6-blocking antibody, whereas GPVI is dispensable for this response, as shown using "knockout" mouse platelets. On the other hand, lamellipodia formation on laminin is completely inhibited in the absence of GPVI, although filopodia formation remains and is presumably mediated via alpha6beta1 Lamellipodia and filopodia formation are inhibited in Syk-deficient platelets, demonstrating a key role for the kinase in signaling downstream of GPVI and integrin alpha6beta1 GPVI was confirmed as a receptor for laminin using surface plasmon resonance spectroscopy and by demonstration of lamellipodia formation on laminin in the presence of collagenase. These results identify GPVI as a novel receptor for laminin and support a model in which integrin alpha6beta1 brings laminin to GPVI, which in turn mediates lamellipodia formation. We speculate that laminin contributes to platelet spreading in vivo through a direct interaction with GPVI.
Collapse
Affiliation(s)
- Osamu Inoue
- Department of Clinical and Laboratory Medicine, Yamanashi Medical University, 1110 Shimokato Tamaho Nakakoma, Yamanashi 409-3898, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Affiliation(s)
- D S Woulfe
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
236
|
Soulet C, Hechler B, Gratacap MP, Plantavid M, Offermanns S, Gachet C, Payrastre B. A differential role of the platelet ADP receptors P2Y1 and P2Y12 in Rac activation. J Thromb Haemost 2005; 3:2296-306. [PMID: 16194206 DOI: 10.1111/j.1538-7836.2005.01588.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The dynamics of the actin cytoskeleton, largely controlled by the Rho family of small GTPases (Rho, Rac and Cdc42), is critical for the regulation of platelet responses such as shape change, adhesion, spreading and aggregation. Here, we investigated the role of adenosine diphosphate (ADP), a major co-activator of platelets, on the activation of Rac. ADP rapidly activated Rac in a dose-dependent manner and independently of GPIIb/IIIa and phosphoinositide 3-kinase. ADP alone, used as a primary agonist, activated Rac and its effector PAK via its P2Y1 receptor, through a G(q)-dependent pathway and independently of P2Y12. The P2Y12 receptor appeared unable to activate the GTPase per se as also observed for the adenosine triphosphate receptor P2X1. Conversely, secreted ADP strongly potentiated Rac activation induced by FcgammaRIIa clustering or TRAP via its P2Y12 receptor, the target of antithrombotic thienopyridines. Stimulation of the alpha(2A)-adrenergic receptor/G(z) pathway by epinephrine was able to replace the P2Y12/G(i)-mediated pathway to amplify Rac activation by FcgammaRIIa or by the thrombin receptor PAR-1. This co-activation appeared necessary to reach a full stimulation of Rac as well as PAK activation and actin polymerization and was blocked by a G-protein betagamma subunits scavenger peptide.
Collapse
Affiliation(s)
- C Soulet
- Inserm U563, Département d'Oncogenèse et signalisation dans les cellules hématopoïétiques, Hôpital Purpan, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
237
|
von Beckerath N, von Beckerath O, Koch W, Eichinger M, Schömig A, Kastrati A. P2Y12 gene H2 haplotype is not associated with increased adenosine diphosphate-induced platelet aggregation after initiation of clopidogrel therapy with a high loading dose. Blood Coagul Fibrinolysis 2005; 16:199-204. [PMID: 15795539 DOI: 10.1097/01.mbc.0000164429.21040.0a] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A large variability in the antiplatelet response to clopidogrel has been consistently reported. Recently, a P2Y12 haplotype was shown to be associated with enhanced adenosine diphosphate (ADP)-induced platelet aggregation in healthy volunteers. The aim of this study was to test in patients (n = 416) scheduled for coronary artery stenting whether P2Y12 haplotype H2 carriage is associated with increased ADP-induced platelet aggregation after administration of a 600 mg loading dose of clopidogrel. Blood was drawn from the arterial sheath at least 2 h after administration of 100 mg aspirin and 600 mg clopidogrel. ADP-induced platelet aggregation was assessed in platelet-rich plasma with light-transmission aggregometry. P2Y12 haplotypes (H1/H2) and P2Y12 C32T genotypes were determined with TaqMan assays. Haplotype combinations and genotypes were not associated with parameters of ADP-induced platelet aggregation after administration of a 600 mg loading dose of clopidogrel. Maximal ADP (5 mumol/l)-induced platelet aggregation was similar in patients carrying haplotype H2 and homozygous carriers of haplotype H1 (43.9 +/- 21.4 versus 43.2 +/- 21.1%, respectively; P = 0.77). Carriage of P2Y12 H2 haplotype does not seem to affect the platelet response to a 600 mg loading dose of clopidogrel in coronary artery disease patients prior to stenting.
Collapse
Affiliation(s)
- Nicolas von Beckerath
- Deutsches Herzzentrum, Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
238
|
Storey RF, May JA, Heptinstall S. Potentiation of platelet aggregation by heparin in human whole blood is attenuated by P2Y12 and P2Y1 antagonists but not aspirin. Thromb Res 2005; 115:301-7. [PMID: 15668190 DOI: 10.1016/j.thromres.2004.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 09/06/2004] [Accepted: 09/07/2004] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Unfractionated heparin (UFH) potentiates platelet aggregation induced by some agonists. P2Y12 and P2Y1 receptors play a major role in amplifying platelet aggregation. We assessed the ability of cangrelor, a selective P2Y12 antagonist, A2P5P, a selective P2Y1 antagonist, and aspirin to block the potentiating effects of heparin. MATERIALS AND METHODS Whole blood from healthy human volunteers was anticoagulated with either hirudin or UFH 10 IU/ml. Some tubes anticoagulated with hirudin also contained UFH 1 or 10 IU/ml. The low-molecular-weight heparin dalteparin was also assessed. Platelet aggregation was performed using whole blood single-platelet counting. Dense granule release was assessed using 14C-5HT-labelled platelets. RESULTS UFH and, to a lesser extent, dalteparin potentiated platelet aggregation induced by ADP, PAF, 5HT, U46619, epinephrine and TRAP in a concentration-dependent manner but inhibited aggregation induced by collagen. Cangrelor effectively opposed the potentiating effects of heparins on sustained aggregation induced by ADP, PAF, 5HT, U46619 and TRAP but had less effect on epinephrine-induced aggregation, whereas A2P5P was more effective at blocking both the initial phase of ADP-induced aggregation and the aggregation response to epinephrine, reflecting the differences in G protein coupling between the agonist receptors. Aspirin had no effect on potentiation by heparin. Heparins did not increase ADP- or TRAP-induced 14C-5HT release. CONCLUSIONS Heparins potentiate platelet responses to ADP and numerous other agonists. This potentiation is attenuated by cangrelor and A2P5P, and is not mediated by increased dense granule release. ADP receptor antagonists but not aspirin may have potential therapeutic benefits in counteracting the pro-thrombotic effects of heparins.
Collapse
Affiliation(s)
- Robert F Storey
- Cardiovascular Research Unit, Clinical Sciences Centre, Northern General Hospital, Sheffield S5 7AU, UK.
| | | | | |
Collapse
|
239
|
Abstract
The current standard of care for the treatment of arterial thrombosis includes anticoagulants and three classes of antiplatelet agents--aspirin, thienopyridines and glycoprotein IIb-IIIa antagonists. Although these drugs have had a significant impact on morbidity and mortality in several patient populations, up to 15% of the high risk patients with acute coronary syndrome continue to suffer from ischemic events. This problem may occur, in part, because the platelets in many patients are non-responsive to aspirin and clopidogrel. Murine models now indicate that platelets are not only responsible for arterial occlusion, they are also involved in the progression of atherosclerotic disease. New opportunities have emerged identifying potential targets and strategies for drug discovery suited to address these deficiencies by more effectively modulating platelet adhesion, thrombus growth, thrombus stability and the pro-inflammatory activity of platelets. In addition, a growing need has emerged for the development of bedside devices capable of bringing personalized medicine to patients being treated with antithrombotic drugs in order to measure the pharmacodynamic activities of new therapies, to assess the activities achieved by combined antithrombotic therapy, and to identify patients that fail to respond.
Collapse
Affiliation(s)
- D R Phillips
- Portola Pharmaceuticals, Inc., San Francisco, CA 94402, USA.
| | | | | | | |
Collapse
|
240
|
|
241
|
Abstract
Platelets play a central role in hemostasis and thrombosis but also in the initiation of atherosclerosis, making platelet receptors and their intracellular signaling pathways important molecular targets for antithrombotic and anti-inflammatory therapy. Historically, much of the knowledge about hemostasis and thrombosis has been derived from patients suffering from bleeding and thrombotic disorders and the identification of the underlying molecular defects. In recent years, the availability of genetically modified mouse strains with defined defects in platelet function and the development of in vivo models to assess platelet-related physiologic and pathophysiologic processes have opened new ways to identify the individual roles and the interplay of platelet proteins in adhesion, activation, aggregation, secretion, and procoagulant activity in vitro and in vivo. This review will summarize key findings made by these approaches and discuss them in the context of human disease.
Collapse
Affiliation(s)
- B Nieswandt
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany.
| | | | | | | |
Collapse
|
242
|
Gachet C. The platelet P2 receptors as molecular targets for old and new antiplatelet drugs. Pharmacol Ther 2005; 108:180-92. [PMID: 15955565 DOI: 10.1016/j.pharmthera.2005.03.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2005] [Accepted: 03/24/2005] [Indexed: 12/15/2022]
Abstract
Platelet activation by ADP and ATP plays a crucial role in haemostasis and thrombosis, and their so-called P2 receptors are potential targets for antithrombotic drugs. The ATP-gated channel P2X1 and the 2 G protein-coupled P2Y1 and P2Y12 ADP receptors selectively contribute to platelet aggregation. The P2Y1 receptor is responsible for ADP-induced shape change and weak and transient aggregation, while the P2Y12 receptor is responsible for the completion and amplification of the response to ADP and to all platelet agonists, including thromboxane A2 (TXA2), thrombin, and collagen. The P2X1 receptor is involved in platelet shape change and in activation by collagen under shear conditions. Due to its central role in the formation and stabilization of a thrombus, the P2Y12 receptor is a well-established target of antithrombotic drugs like ticlopidine or clopidogrel, which have proved efficacy in many clinical trials and experimental models of thrombosis. Competitive P2Y12 antagonists have also been shown to be effective in experimental thrombosis as well as in several clinical trials. Studies in P2Y1 and P2X1 knockout mice and experimental thrombosis models using selective P2Y1 and P2X1 antagonists have shown that, depending on the conditions, these receptors could also be potential targets for new antithrombotic drugs.
Collapse
Affiliation(s)
- Christian Gachet
- INSERM U.311, Etablissement Français du Sang-Alsace, 10 rue Spielmann, B.P. No. 36, 67065 Strasbourg Cedex, France.
| |
Collapse
|
243
|
Tolhurst G, Vial C, Léon C, Gachet C, Evans RJ, Mahaut-Smith MP. Interplay between P2Y(1), P2Y(12), and P2X(1) receptors in the activation of megakaryocyte cation influx currents by ADP: evidence that the primary megakaryocyte represents a fully functional model of platelet P2 receptor signaling. Blood 2005; 106:1644-51. [PMID: 15914557 DOI: 10.1182/blood-2005-02-0725] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The difficulty of conducting electrophysiologic recordings from the platelet has restricted investigations into the role of ion channels in thrombosis and hemostasis. We now demonstrate that the well-established synergy between P2Y(1) and P2Y(12) receptors during adenosine diphosphate (ADP)-dependent activation of the platelet alpha(IIb)beta(3) integrin also exists in murine marrow megakaryocytes, further supporting the progenitor cell as a bona fide model of platelet P2 receptor signaling. In patch clamp recordings, ADP (30 microM) stimulated a transient inward current at -70 mV, which was carried by Na(+) and Ca(2+) and was amplified by phenylarsine oxide, a potentiator of certain transient receptor potential (TRP) ion channels by phosphatidylinositol 4,5-bisphosphate depletion. This initial current decayed to a sustained phase, upon which repetitive transient inward cation currents with pre-dominantly P2X(1)-like kinetics were super-imposed. Abolishing P2X(1)-receptor activity prevented most of the repetitive currents, consistent with their activation by secreted adenosine triphosphate (ATP). Recordings in P2Y(1)-receptor-deficient megakaryocytes demonstrated an essential requirement of this receptor for activation of all ADP-evoked inward currents. However, P2Y(12) receptors, through the activation of PI3-kinase, played a synergistic role in both P2Y(1) and P2X(1)-receptor-dependent currents. Thus, direct stimulation of P2Y(1) and P2Y(12) receptors, together with autocrine P2X(1) activation, is responsible for the activation of nonselective cation currents by the platelet agonist ADP.
Collapse
Affiliation(s)
- Gwen Tolhurst
- Department of Physiology, University of Cambridge, Cambridge, CB2 3EG, United Kingdom
| | | | | | | | | | | |
Collapse
|
244
|
Quinton TM, Kim S, Jin J, Kunapuli SP. Lipid rafts are required in Galpha(i) signaling downstream of the P2Y12 receptor during ADP-mediated platelet activation. J Thromb Haemost 2005; 3:1036-41. [PMID: 15869601 DOI: 10.1111/j.1538-7836.2005.01325.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
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.
Collapse
Affiliation(s)
- T M Quinton
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | |
Collapse
|
245
|
Campus F, Lova P, Bertoni A, Sinigaglia F, Balduini C, Torti M. Thrombopoietin complements G(i)- but not G(q)-dependent pathways for integrin {alpha}(IIb){beta}(3) activation and platelet aggregation. J Biol Chem 2005; 280:24386-95. [PMID: 15863506 DOI: 10.1074/jbc.m501174200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
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.
Collapse
MESH Headings
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
- Adenosine Diphosphate/chemistry
- Androstadienes/pharmacology
- Antibodies, Monoclonal/chemistry
- Blood Platelets/cytology
- 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/genetics
- GTP-Binding Protein alpha Subunits, Gi-Go/physiology
- GTP-Binding Protein alpha Subunits, Gq-G11/genetics
- GTP-Binding Protein alpha Subunits, Gq-G11/physiology
- Humans
- Morpholines/pharmacology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoproteins/chemistry
- Phosphoproteins/metabolism
- Platelet Activation
- Platelet Aggregation
- Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
- Protein Binding
- Proto-Oncogene Proteins/metabolism
- Receptors, Cytokine/metabolism
- Receptors, Purinergic P2/metabolism
- Receptors, Purinergic P2Y1
- Receptors, Thrombopoietin
- Thrombopoietin/chemistry
- Thrombopoietin/genetics
- Thrombopoietin/metabolism
- Thrombopoietin/physiology
- Thromboxane A2/metabolism
- Thromboxanes/chemistry
- Time Factors
- Type C Phospholipases/metabolism
- Wortmannin
- rap GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Francesca Campus
- Department of Biochemistry, University of Pavia, via Bassi 21, 27100 Pavia
| | | | | | | | | | | |
Collapse
|
246
|
Léon C, Freund M, Latchoumanin O, Farret A, Petit P, Cazenave JP, Gachet C. The P2Y(1) receptor is involved in the maintenance of glucose homeostasis and in insulin secretion in mice. Purinergic Signal 2005; 1:145-51. [PMID: 18404499 PMCID: PMC2096536 DOI: 10.1007/s11302-005-6209-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Accepted: 10/18/2004] [Indexed: 11/21/2022] Open
Abstract
Pancreatic β cells express several P2 receptors including P2Y1 and the modulation of insulin secretion by extracellular nucleotides has suggested that these receptors may contribute to the regulation of glucose homeostasis. To determine whether the P2Y1 receptor is involved in this process, we performed studies in P2Y1 mice. In baseline conditions, P2Y1-mice exhibited a 15% increase in glycemia and a 40% increase in insulinemia, associated with a 10% increase in body weight, pointing to a role of the P2Y1 receptor in the control of glucose metabolism. Dynamic experiments further showed that P2Y1-mice exhibited a tendency to glucose intolerance. These features were associated with a decrease in the plasma levels of free fatty acid and triglycerides. When fed a lipids and sucrose enriched diet for 15 weeks, the two genotypes no longer displayed any significant differences. To determine whether the P2Y1 receptor was directly involved in the control of insulin secretion, experiments were carried out in isolated Langerhans islets. In the presence of high concentrations of glucose, insulin secretion was significantly greater in islets from P2Y1-mice. Altogether, these results show that the P2Y1 receptor plays a physiological role in the maintenance of glucose homeostasis at least in part by regulating insulin secretion.
Collapse
Affiliation(s)
- Catherine Léon
- Institut National de la Santé et de la Recherche Médicale U.311, Etablissement Français du Sang-Alsace, Strasbourg Cedex, France
| | | | | | | | | | | | | |
Collapse
|
247
|
Robson SC, Sévigny J, Imai M, Guckelberger O, Enjyoji K. Thromboregulatory potential of endothelial CD39/nucleoside triphosphate diphosphohydrolase: modulation of purinergic signalling in platelets. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.4.2.155] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
248
|
|
249
|
Görgen M, Turatti K, Medeiros AR, Buffon A, Bonan CD, Sarkis JJF, Pereira GS. Aqueous extract of Ilex paraguariensis decreases nucleotide hydrolysis in rat blood serum. JOURNAL OF ETHNOPHARMACOLOGY 2005; 97:73-77. [PMID: 15652278 DOI: 10.1016/j.jep.2004.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2004] [Revised: 09/24/2004] [Accepted: 10/18/2004] [Indexed: 05/24/2023]
Abstract
Mate is a xanthine-containing beverage, which is prepared as an infusion of the dried and ground leaves of Ilex paraguariensis St. Hil. (Aquifoliacea). Previous reports have shown that Ilex paraguariensis has the highest levels of caffeine and theobromine when compared to other Ilex species. Furthermore, mate is able to interfere in the circulatory system, acting as a diuretic and hypotensive agent. Many processes of vascular injury result in the release of adenine nucleotides, which exert a variety of effects. Nucleoside 5' tri- and diphosphates may be hydrolyzed by members of the ecto-nucleoside triphosphate diphosphohydrolase (E-NTPDase) family. The synchronic action of a NTPDase and a 5'-nucleotidase promotes the catabolism of ATP to adenosine, which is able to control the extracellular nucleotides/nucleosides ratio. The chronic ingestion of aqueous extract of Ilex paraguariensis by rats during 15 days significantly decreased ATP (55%), ADP (50%) and AMP (40%) hydrolysis in blood serum. These results suggest changes in the balance of purine levels induced by Ilex paraguariensis ingestion. Considering the potential effects of Ilex paraguariensis in the circulatory system, these results may be relevant since NTPDases are a novel drug target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Milena Görgen
- Laboratório de Bioquímica, UNIVATES Centro Universitário, Lajeado, Rua Avelino Tallini, 171 CEP: 95900-000 Caixa Posta 155, Lajeado, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
250
|
Abstract
The platelet surface membrane possesses three P2 receptors activated by extracellular adenosine nucleotides; one member of the ionotropic receptor family (P2X(1)) and two members of the G-protein-coupled receptor family (P2Y(1) and P2Y(12)). P2Y(1) and P2Y(12) receptors have firmly established roles in platelet activation during thrombosis and haemostasis, whereas the importance of the P2X(1) receptor has been more controversial. However, recent studies have demonstrated that P2X(1) receptors can generate significant functional platelet responses alone and in synergy with other receptor pathways. In addition, studies in transgenic animals indicate an important role for P2X(1) receptors in platelet activation, particularly under conditions of shear stress and thus during arterial thrombosis. This review discusses the background behind discovery of P2X(1) receptors in platelets and their precursor cell, the megakaryocyte, and how signalling via these ion channels may participate in platelet activation.
Collapse
Affiliation(s)
- Martyn P Mahaut-Smith
- Department of Physiology, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | | | | |
Collapse
|