Hamostaseologie 2020; 40(02): 145-152
DOI: 10.1055/a-1113-0711
Review Article
Georg Thieme Verlag KG Stuttgart · New York

Platelet Purinergic Receptors in Thrombosis and Inflammation

Christian Gachet
1   Etablissement Français du Sang (EFS) Grand Est, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg Cedex, France
,
Beatrice Hechler
1   Etablissement Français du Sang (EFS) Grand Est, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg Cedex, France
› Author Affiliations
Further Information

Address for correspondence

Christian Gachet, MD, PhD
Etablissement Français du Sang-Grand Est
INSERM UMR_S1255, 10, rue Spielmann, BP 36, F-67065 Strasbourg Cedex
France   

Publication History

12 November 2019

23 January 2020

Publication Date:
28 May 2020 (online)

 

Abstract

It took approximately 40 years from the seminal identification of adenosine diphosphate (ADP) as the factor R, an agent derived from red blood cells inducing platelet adhesion to glass, to the completion of the repertoire of its receptors on platelets and its importance in haemostasis and thrombosis. ADP, either derived from red blood cells or released by platelets themselves, stimulates platelets via two G protein-coupled receptors, P2Y1 and P2Y12. In addition, adenosine triphosphate, also contained in the platelet dense granules, activates the P2X1 cation channel. Each of these receptors plays a specific role during platelet activation and aggregation, with relevance to haemostasis, thrombosis and various inflammatory processes where platelets are involved including chronic responses such as atherosclerosis or acute responses such as sepsis, endotoxaemia or allergic asthma. Finally, platelets also express P2Y14, a receptor activated by released uridine diphosphate glucose. Although devoid of any known role in haemostasis, this receptor seems to play a specific role in neutrophil chemotaxis.


#

Introduction

Sixty years ago now, adenosine diphosphate (ADP) was identified as a factor released from erythrocytes which influenced platelet adhesiveness to glass[1] and induced platelet aggregation.[2] The crucial role of ADP as a mediator of platelet activation was rapidly recognized in the physiological process of haemostasis and in the development and extension of arterial thrombosis.[3]

ADP and adenosine triphosphate (ATP) are stored at a very high concentration in platelet dense granules and released upon activation of platelets by strong agonists such as thrombin and/or collagen. Released ADP is an essential secondary agonist, which amplifies most of the platelet responses and contributes to the stabilisation of the thrombus.[4] Platelet stimulation by ADP leads to a transient increase in free cytoplasmic Ca2+ and inhibition of adenylyl cyclase activity. In addition, ATP induces an extremely rapid influx of Ca2+ from the extracellular medium associated to platelet shape change. We know for years now that three different P2 receptors mediate these effects of adenine nucleotides on platelets: two G protein-coupled receptors stimulated by ADP, P2Y1 and P2Y12, and the P2X1 cation channel activated by ATP. Each of these receptors plays a specific role during platelet activation and aggregation, with relevance to haemostasis, thrombosis and various inflammatory processes where platelets are involved including chronic responses such as atherosclerosis or acute responses such as sepsis, endotoxaemia or allergic asthma. Platelets also express the P2Y14 receptor activated by uridine diphosphate (UDP) and UDP-glucose but its role, if any, remains elusive.[5] The role of the P2 receptors in platelet activation, thrombus formation and inflammation has been presented and discussed in many reviews by us and by others that we cannot cite all for reasons of space. In this short overview, we will only briefly highlight several key points.


#

The Platelet P2Y1 Receptor

The P2Y1 receptor is broadly expressed in many cells and tissues. Its presence and role in platelets were established by the detection of mRNA in megakaryoblastic cell lines and by pharmacological studies using selective P2Y1 antagonists.[6] ADP is the preferred natural agonist of the P2Y1 receptor, while ATP behaves as an antagonist in platelets[7] or as a poor partial agonist in heterologous transfected or reconstituted systems, depending on the receptor density.[8] The P2Y1 receptor is coupled to Gq and Ca2+ signalling. The changes in cytosolic Ca2+ concentration support the activation of the calcium and diacylglycerol-regulated guanine-nucleotide exchange factor 1 (CalDAG-GEFI), resulting in rapid and reversible activation of the small RAP1 GTPase and integrin αIIbβ3, responsible for platelet shape change and weak and transient aggregation in response to ADP.[9] Platelets express approximately 150 P2Y1 receptors per cell,[10] which is very low and probably explains why the signal induced by P2Y1 activation is weak as compared with the signals of other Gq-coupled receptors. To date, no P2Y1-deficient patients have been identified.


#

The Platelet P2Y12 Receptor

The P2Y12 receptor, cloned in 2001,[11] [12] is responsible for completion of the platelet aggregation initiated by the P2Y1 receptor in response to ADP,[13] as well as for the role played by ADP in the amplification of aggregation, secretion and stabilisation of platelet aggregates induced by strong agonists such as thrombin, collagen or TxA2.[4] [14] It is activated by ADP, while ATP and a wide range of its triphosphate analogues behave as antagonists.[15] Platelets express approximately 400 copies of the P2Y12 receptor per cell. Owing its central role in the amplification of platelet responses to any stimulus, it is the molecular target of potent antiplatelet drugs including the thienopyridine compounds clopidogrel and prasugrel, which are prodrugs, and the direct antagonists ticagrelor and cangrelor.[16] [17] The tissue distribution of this receptor was long thought to be restricted to platelets and sub-regions of the brain. Further studies later revealed its expression and functions in microglial cells, vascular smooth muscle cells (VSMCs), dendritic cells (DCs), macrophages and as yet unspecified leukocytes and also bladder smooth muscle cells.[18] [19] [20] In terms of signalling, the P2Y12 receptor activates a Gαi2 G-protein subtype, responsible for the activation of two phosphoinositide 3-kinase (PI3-K) isoforms (PI3-K p110β and p110 gamma) that regulate αIIbβ3 activation via activation of the serine–threonine protein kinase B/Akt (PKB/Akt) and the small GTPase RAP1 and the downward regulation of the RAP1 RAP-GAP RASA3 inhibitor.[21] [22] Gαi2 also inhibits platelet adenylyl cyclase, which facilitates platelet activation.

Several patients have been identified with congenital P2Y12 receptor defects associated with quantitative or qualitative abnormalities of the receptor and responsible for deficiencies of ADP-induced platelet activation, leading to mild to severe bleeding diathesis which underscores the importance of this receptor in haemostasis.[16] [23]


#

The Platelet P2X1 Receptor

The third member of the platelet P2 receptor panoply is the ligand-gated cation channel P2X1, responsible for a fast Ca2+ entry induced by ATP. Pharmacological studies using specific P2X1 ligands and P2X1 −/− mice have shown that this receptor triggers transient shape change without causing platelet aggregation in response to ATP[24] and participates in collagen- and shear-induced aggregation. No P2X1-deficient patients have been clearly identified to date. A mutation in the P2X1 sequence has been reported in a patient with a severe bleeding disorder, but without firm demonstration of a causal link between the mutation and the bleeding tendency.[25] [26]


#

The Platelet P2 Receptors in Thrombosis

So far, only the P2Y12 receptor is an established target for antithrombotic drugs in clinical use. The P2Y1 and P2X1 receptors are also involved in experimental thrombosis and are at a preclinical stage of evaluation as potential targets for new antiplatelet agents.


#

The Platelet P2Y12 Receptor in Thrombosis

The cornerstone of treatment of ischemic coronary syndromes is the dual antiplatelet therapy using aspirin and P2Y12-targeting drugs.[17] Indeed, the central role of the P2Y12 receptor in platelet activation and the growth and stabilisation of a thrombus makes it a very important molecular target for antithrombotic agents.[16] [18] It is so far the only P2 receptor subtype to be an established target for antiplatelet drugs in clinical use and the major target to treat all kinds of arterial ischemic diseases. Active metabolites of the orally administered thienopyridine prodrugs (clopidogrel and prasugrel) covalently bind to the P2Y12 receptor while the direct-acting P2Y12 antagonists, ticagrelor, which is an oral antagonist, and cangrelor, an intravenous direct antagonist, reversibly bind the receptor and inhibit ADP binding. All these drugs are under clinical use for the treatment and prevention of thrombotic events in acute coronary syndromes. However, one limitation of targeting the P2Y12 receptor, whatever the drug, relates to the bleeding risk, which increases with the degree of inhibition of P2Y12-dependent platelet functions.[27] The crystal structure of P2Y12 revealed how this receptor behaves when it binds agonists and antagonists. This should provide valuable insights into the development of improved P2Y12 antagonists.[28] [29]


#

The Platelet P2Y1 Receptor in Thrombosis

Although the P2Y1 receptor plays only a modest role in platelet activation, it plays a central role in experimental arterial thrombosis. This was shown using P2Y1 −/− mice in various models including systemic thromboembolism either induced by infusion of a mixture of collagen and adrenaline[30] [31] or tissue factor,[32] or localised thrombosis after ferric chloride- or laser-induced injury of mesenteric arteries.[33] Among selective P2Y1 receptor antagonists, MRS2500 has so far the highest affinity for P2Y1 and is able to inhibit thrombosis in treated animals.[34] However, the limited bioavailability of the compound prevents its use in long-term treatment, pointing to the need of developing novel P2Y1 receptor antagonists with an improved pharmacokinetic profile. The resolution of the X-ray crystal structure of the P2Y1 receptor[35] should open up new possibilities in this field. Of note, inhibition of the P2Y1 receptor results in a moderate prolongation of the bleeding time in mice, which is a definite safety advantage over inhibition of the P2Y12 receptor. However, so far, no candidate drug has really emerged from any pharmaceutical company involved in antithrombotic drug development.


#

The Platelet P2X1 Receptor in Thrombosis

The generation of P2X1 −/− mice helped to reveal the important role of this receptor in arterial thrombosis. P2X1 −/− mice display resistance to systemic thromboembolism induced by injection of a mixture of collagen and adrenaline and to localised arterial thrombosis triggered by laser-induced injury of mesenteric arteries.[24] These mice display also resistance to tissue factor-induced systemic thromboembolism, i.e., in a thrombin-dependent system (unpublished data). Interestingly, P2X1 −/− mice have no increase in their bleeding time compared with wild-type animals, indicating that they display normal haemostasis.[24] This receptor is therefore a potential new target for safe antiplatelet agents. However, due to the limited bioavailability and selectivity of currently existing P2X1 antagonists, the development of novel selective P2X1 antagonists is warranted for the preclinical evaluation of this receptor as a target for novel antiplatelet agents.


#

The Platelet P2 Receptors in Inflammation

Platelets play an important role in modulating inflammatory responses through the release of inflammatory mediators or compounds with trophic activity and exposure of P-selectin, CD40 and CD40 ligand (CD40L). These molecules allow interaction of platelets with leukocytes and their subsequent activation with the release of a range of inflammatory cytokines and exposure of tissue factor.[36] Therefore, in addition to acting as antithrombotic drugs, antagonists and inhibitors of the platelet P2 receptors have anti-inflammatory effects, which might be relevant to various diseases such as atherosclerosis, restenosis, endotoxaemia and sepsis, or allergic asthma. In addition, besides platelets, P2Y1, P2Y12 and P2X1 receptors, expressed by cells of the immune system and by vascular cells, are also directly involved in the modulation of inflammation and in immune responses during thrombus formation, independently of platelet-related processes[37] ([Table 1]).

Table 1

Role of the P2Y1, P2Y12, P2X1 and P2Y14 receptors in inflammatory processes and related diseases

Inflammatory disease

Cell type

Reference

Atherosclerosis

P2Y12

Platelets

Li et al (2012)[40]; West et al (2014)[41]

VSMCs

Li et al (2012)[40]; West et al (2014)[41]

P2Y1

Endothelial cells

Hechler et al (2008)[51]; Zerr et al (2011)[50]

Restenosis

P2Y12

Platelets

Evans et al (2009)[31]

Transplant atherosclerosis

P2Y12

VSMCs

Abele et al (2009)[42]; Harada et al (2011)[43]

Leukocytes

Abele et al (2009)[42]; Harada et al (2011)[43]

Intimal hyperplasia

P2Y1

Platelets

Liu et al (2015)[52]

Macrophages

Liu et al (2015)[52]; Marques-da-Silva et al (2011)[53]

Endotoxaemia/sepsis

P2Y12

Platelets

Hagiwara et al (2011)[47]; Liverani et al (2016)[48]

P2X1

Neutrophils

Lecut et al (2012)[56]; Maître et al (2015)[57]

P2Y14

Platelets

Sesma et al (2016)[59]; Amison et al (2017)[60]

Transfusion-related acute lung injury

P2X1

Monocytes/macrophages

El Mdawar et al (2019)[58]

Asthma

P2Y12

Platelets

Paruchuri et al (2009)[44]; Cattaneo (2015)[45]

P2Y1

Platelets

Amison et al (2015)[46]

Abbreviation: VSMCs, vascular smooth muscle cells.



#

The P2Y12 Receptor

Studies in mice or patients receiving clopidogrel have highlighted the contribution of the platelet P2Y12 receptor to P-selectin and CD40L exposure, formation of platelet–leukocyte aggregates, tissue factor exposure and release of various inflammatory mediators (tumour necrosis factor-α, C-reactive protein).[38] These inflammatory events might be relevant for the role played by the platelet P2Y12 receptor in restenosis,[39] atherosclerosis[40] [41] and transplant atherosclerosis. An additional contribution from the P2Y12 receptor of VSMCs and leukocytes, including DCs, possibly through P2Y12-dependent migration properties has been suggested.[42] [43] In the context of allergic asthma, in which platelet activation is required for the recruitment of inflammatory cells to the lungs and remodelling of the airway wall, divergent results have been reported concerning the role of the platelet P2Y12 receptor. The pro-asthmatic action of leukotriene LTE4 in mice could require the P2Y12 receptor through a mechanism yet to be identified.[44] Further evidence of the involvement of the P2Y12 platelet receptor in asthma is provided by clinical observations of a large family-based asthma cohort, in which P2Y12 receptor variants were associated with altered lung function, while prasugrel tended to decrease the bronchial hyper-reactivity to mannitol in patients with allergic asthma.[45] In contrast, Amison et al reported that the platelet P2Y12 receptor is dispensable in allergic inflammation in ovalbumin-challenged mice.[46] The platelet P2Y12 receptor contributes also to the deleterious pro-inflammatory role of platelets in enhancing the pulmonary neutrophil infiltration and exacerbating tissue damage during lipopolysaccharide (LPS)-induced endotoxemia.[47] Concerning the potential role of the platelet P2Y12 receptor in sepsis, here also, contradictory results have been reported. On the one hand, Liverani et al reported a central role of P2Y12,[48] while on the other hand our group reported that inhibition of the P2Y12 receptor with clopidogrel or platelet P2Y12 receptor deficiency (unpublished data) does not protect mice from septic shock. Further studies are required to clarify all these discrepancies, most probably related to differences in models, administered drugs and/or the use of receptor-deficient mice, to elucidate the contribution of the P2Y12 receptor in various settings. The perspective could be to extend the use of P2Y12 targeting drugs beyond their current indications.


#

The P2Y1 Receptor

The platelet P2Y1 receptor contributes to platelet P-selectin exposure and the formation of platelet–leukocyte conjugates resulting in leukocyte activation and tissue factor exposure.[49] These P2Y1 receptor-mediated events might be relevant for the platelet-dependent recruitment of leukocytes to the lung tissue in the case of inflammation of the airways in allergic mice,[46] although the contribution of the P2Y1 receptors from other cell types such as leukocytes or endothelial cells cannot be excluded. Indeed, in an experimental model of acute vascular inflammation, the endothelial P2Y1 receptor plays an important role in the upregulation of adhesion molecules (P-selectin, VCAM-1, ICAM-1 and the recruitment of leukocytes at the vascular wall).[50] The endothelial P2Y1 receptor also contributes to development of atherosclerosis, a chronic inflammatory process, in ApoE−/− mice.[51] The P2Y1 receptor has also been shown to be present on VSMCs, where it contributes to their proliferation and migration in vitro. This could be relevant for the intimal hyperplasia observed in a vein graft model in mice.[52] In this case, a contribution from the P2Y1 receptors present on platelets and macrophages cannot be excluded. Macrophages would indeed appear to be required, as their depletion abrogated hyperplasia,[52] while data indicated a role of the P2Y1 receptor in the phagocytic and migration activity of macrophages.[53] Altogether, these pieces of evidence suggest that the P2Y1 receptor could represent an attractive and original target for drugs with multiple sites of action, to treat atherothrombosis and, possibly, other inflammatory diseases. Again, the main problem is the lack of good drug candidates to further evaluate this receptor as a potential target for new drug candidates.


#

The P2X1 Receptor

The platelet P2X1 receptor, along with the neutrophil P2X1 receptor, may contribute to thrombus formation in a context of inflammation as highlighted in a specific model of neutrophil-dependent thrombosis of cremasteric arterioles triggered by laser injury. The neutrophil P2X1 receptor appeared essential for their recruitment at the site of vessel injury and subsequent fibrin production and thrombus formation.[54] Very recently, a role of the platelet P2X1 receptor in the enhancement of FcγRIIa-induced Ca2+ increases and functional responses has been reported, which may be relevant for in vivo platelet-dependent immune responses.[55] The P2X1 receptor from neutrophils plays also an important part in facilitating the neutrophil chemotaxis induced by various chemoattractants, possibly by favouring contraction and retraction of the trailing uropod.[56] Accordingly, the P2X1 receptor from neutrophil, or from monocyte/macrophage, displays an important role in inflammatory diseases leading to organ damage, such as endotoxemia[57] or transfusion-related acute lung injury (TRALI).[58] Thus, similarly to P2Y1, the P2X1 receptor could constitute an attractive target for new therapeutics not only in thrombosis but also in inflammatory processes. Here also, the lack of potent and selective compounds with favourable bioavailability hampers the further evaluation of the potential of this receptor as a valuable target for new drugs.


#

The P2Y14 Receptor

As already mentioned, platelets express the P2Y14 receptor but its function in platelet physiology is unknown and no modulation of thrombosis has been detected using P2Y14 −/− mice in various models of arterial thrombosis (unpublished data). However, using P2Y14 antagonists, separate studies have reported a role of this receptor in leukocyte recruitment in vitro and in vivo. Pulmonary neutrophil recruitment induced by intranasal LPS administration and LPS-induced thrombocytopenia were inhibited in mice administered with a P2Y14 antagonist.[59] [60] In addition, the stimulation of platelets with selective P2Y14 agonists (UDP-glucose, MRS2690) resulted in significant platelet-dependent neutrophil chemotaxis toward macrophage-derived chemokine (CCL22), in vitro. Similarly, the P2Y1 receptor seems to play a specific role in platelet-dependent neutrophil chemotaxis.[60]


#

Conclusions

Each of the platelet receptors for extracellular adenine nucleotides plays a specific role in platelet functions and arterial thrombosis. Each of these receptors also contributes to the development of various processes related to acute or chronic inflammation. The respective contributions of the platelet receptors and those of other cell types are not yet well unravelled. Undoubtedly, the generation of mice with a specific tissue deletion of these P2 receptor subtypes will help to define their specific functions under various pathological conditions. In addition, the development of novel P2X1 and P2Y1 receptor antagonists with an improved pharmacokinetic profile should clearly open up new possibilities in this field. The perspective is that these P2 receptors could also represent potential therapeutic targets for the treatment of inflammatory diseases.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Disclosures

None.


  • References

  • 1 Gaarder A, Jonsen J, Laland S, Hellem A, Owren PA. Adenosine diphosphate in red cells as a factor in the adhesiveness of human blood platelets. Nature 1961; 192: 531-532
  • 2 Born GVR. Aggregation of blood platelets by adenosine diphosphate and its reversal. Nature 1962; 194: 927-929
  • 3 Maffrand JP, Bernat A, Delebassée D, Defreyn G, Cazenave JP, Gordon JL. ADP plays a key role in thrombogenesis in rats. Thromb Haemost 1988; 59 (02) 225-230
  • 4 Cattaneo M, Canciani MT, Lecchi A. , et al. Released adenosine diphosphate stabilizes thrombin-induced human platelet aggregates. Blood 1990; 75 (05) 1081-1086
  • 5 Dovlatova N, Wijeyeratne YD, Fox SC. , et al. Detection of P2Y(14) protein in platelets and investigation of the role of P2Y(14) in platelet function in comparison with the EP(3) receptor. Thromb Haemost 2008; 100 (02) 261-270
  • 6 Hechler B, Léon C, Vial C. , et al. The P2Y1 receptor is necessary for adenosine 5′-diphosphate-induced platelet aggregation. Blood 1998; 92 (01) 152-159
  • 7 Léon C, Hechler B, Vial C, Leray C, Cazenave JP, Gachet C. The P2Y1 receptor is an ADP receptor antagonized by ATP and expressed in platelets and megakaryoblastic cells. FEBS Lett 1997; 403 (01) 26-30
  • 8 Waldo GL, Harden TK. Agonist binding and Gq-stimulating activities of the purified human P2Y1 receptor. Mol Pharmacol 2004; 65 (02) 426-436
  • 9 Crittenden JR, Bergmeier W, Zhang Y. , et al. CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation. Nat Med 2004; 10 (09) 982-986
  • 10 Gachet C. Regulation of platelet functions by P2 receptors. Annu Rev Pharmacol Toxicol 2006; 46: 277-300
  • 11 Hollopeter G, Jantzen HM, Vincent D. , et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 2001; 409 (6817): 202-207
  • 12 Zhang FL, Luo L, Gustafson E. , et al. ADP is the cognate ligand for the orphan G protein-coupled receptor SP1999. J Biol Chem 2001; 276 (11) 8608-8615
  • 13 Hechler B, Eckly A, Ohlmann P, Cazenave JP, Gachet C. The P2Y1 receptor, necessary but not sufficient to support full ADP-induced platelet aggregation, is not the target of the drug clopidogrel. Br J Haematol 1998; 103 (03) 858-866
  • 14 Trumel C, Payrastre B, Plantavid M. , et al. A key role of adenosine diphosphate in the irreversible platelet aggregation induced by the PAR1-activating peptide through the late activation of phosphoinositide 3-kinase. Blood 1999; 94 (12) 4156-4165
  • 15 Kauffenstein G, Hechler B, Cazenave JP, Gachet C. Adenine triphosphate nucleotides are antagonists at the P2Y receptor. J Thromb Haemost 2004; 2 (11) 1980-1988
  • 16 Cattaneo M. The platelet P2Y12 receptor for adenosine diphosphate: congenital and drug-induced defects. Blood 2011; 117 (07) 2102-2112
  • 17 Gremmel T, Michelson AD, Frelinger III AL, Bhatt DL. Novel aspects of antiplatelet therapy in cardiovascular disease. Res Pract Thromb Haemost 2018; 2 (03) 439-449
  • 18 Gachet C. P2Y(12) receptors in platelets and other hematopoietic and non-hematopoietic cells. Purinergic Signal 2012; 8 (03) 609-619
  • 19 Hechler B, Gachet C. Purinergic receptors in thrombosis and inflammation. Arterioscler Thromb Vasc Biol 2015; 35 (11) 2307-2315
  • 20 Hao Y, Wang L, Chen H. , et al. Targetable purinergic receptors P2Y12 and A2b antagonistically regulate bladder function. JCI Insight 2019; 4 (16) 122112
  • 21 Gratacap MP, Guillermet-Guibert J, Martin V. , et al. Regulation and roles of PI3Kβ, a major actor in platelet signaling and functions. Adv Enzyme Regul 2011; 51 (01) 106-116
  • 22 Stefanini L, Paul DS, Robledo RF. , et al. RASA3 is a critical inhibitor of RAP1-dependent platelet activation. J Clin Invest 2015; 125 (04) 1419-1432
  • 23 Lecchi A, Femia EA, Paoletta S. , et al. Inherited dysfunctional platelet P2Y12 receptor mutations associated with bleeding disorders. Hamostaseologie 2016; 36 (04) 279-283
  • 24 Hechler B, Lenain N, Marchese P. , et al. A role of the fast ATP-gated P2 × 1 cation channel in thrombosis of small arteries in vivo. J Exp Med 2003; 198 (04) 661-667
  • 25 Cattaneo M. The P2 receptors and congenital platelet function defects. Semin Thromb Hemost 2005; 31 (02) 168-173
  • 26 Oury C, Toth-Zsamboki E, Van Geet C. , et al. A natural dominant negative P2 × 1 receptor due to deletion of a single amino acid residue. J Biol Chem 2000; 275 (30) 22611-22614
  • 27 Gachet C. Antiplatelet drugs: which targets for which treatments?. J Thromb Haemost 2015; 13 (Suppl. 01) S313-S322
  • 28 Zhang J, Zhang K, Gao ZG. , et al. Agonist-bound structure of the human P2Y12 receptor. Nature 2014; 509 (7498): 119-122
  • 29 Zhang K, Zhang J, Gao ZG. , et al. Structure of the human P2Y12 receptor in complex with an antithrombotic drug. Nature 2014; 509 (7498): 115-118
  • 30 Fabre JE, Nguyen M, Latour A. , et al. Decreased platelet aggregation, increased bleeding time and resistance to thromboembolism in P2Y1-deficient mice. Nat Med 1999; 5 (10) 1199-1202
  • 31 Léon C, Hechler B, Freund M. , et al. Defective platelet aggregation and increased resistance to thrombosis in purinergic P2Y(1) receptor-null mice. J Clin Invest 1999; 104 (12) 1731-1737
  • 32 Léon C, Freund M, Ravanat C, Baurand A, Cazenave JP, Gachet C. Key role of the P2Y(1) receptor in tissue factor-induced thrombin-dependent acute thromboembolism: studies in P2Y(1)-knockout mice and mice treated with a P2Y(1) antagonist. Circulation 2001; 103 (05) 718-723
  • 33 Lenain N, Freund M, Léon C, Cazenave JP, Gachet C. Inhibition of localized thrombosis in P2Y1-deficient mice and rodents treated with MRS2179, a P2Y1 receptor antagonist. J Thromb Haemost 2003; 1 (06) 1144-1149
  • 34 Hechler B, Nonne C, Roh EJ. , et al. MRS2500 [2-iodo-N6-methyl-(N)-methanocarba-2′-deoxyadenosine-3′,5′-bisphosphate], a potent, selective, and stable antagonist of the platelet P2Y1 receptor with strong antithrombotic activity in mice. J Pharmacol Exp Ther 2006; 316 (02) 556-563
  • 35 Zhang D, Gao ZG, Zhang K. , et al. Two disparate ligand-binding sites in the human P2Y1 receptor. Nature 2015; 520 (7547): 317-321
  • 36 Morrell CN, Aggrey AA, Chapman LM, Modjeski KL. Emerging roles for platelets as immune and inflammatory cells. Blood 2014; 123 (18) 2759-2767
  • 37 Idzko M, Ferrari D, Eltzschig HK. Nucleotide signalling during inflammation. Nature 2014; 509 (7500): 310-317
  • 38 Steinhubl SR, Badimon JJ, Bhatt DL, Herbert JM, Lüscher TF. Clinical evidence for anti-inflammatory effects of antiplatelet therapy in patients with atherothrombotic disease. Vasc Med 2007; 12 (02) 113-122
  • 39 Evans DJ, Jackman LE, Chamberlain J. , et al. Platelet P2Y(12) receptor influences the vessel wall response to arterial injury and thrombosis. Circulation 2009; 119 (01) 116-122
  • 40 Li D, Wang Y, Zhang L. , et al. Roles of purinergic receptor P2Y, G protein-coupled 12 in the development of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012; 32 (08) e81-e89
  • 41 West LE, Steiner T, Judge HM, Francis SE, Storey RF. Vessel wall, not platelet, P2Y12 potentiates early atherogenesis. Cardiovasc Res 2014; 102 (03) 429-435
  • 42 Abele S, Spriewald BM, Ramsperger-Gleixner M. , et al. Attenuation of transplant arteriosclerosis with clopidogrel is associated with a reduction of infiltrating dendritic cells and macrophages in murine aortic allografts. Transplantation 2009; 87 (02) 207-216
  • 43 Harada K, Matsumoto Y, Umemura K. Adenosine diphosphate receptor P2Y12-mediated migration of host smooth muscle-like cells and leukocytes in the development of transplant arteriosclerosis. Transplantation 2011; 92 (02) 148-154
  • 44 Paruchuri S, Tashimo H, Feng C. , et al. Leukotriene E4-induced pulmonary inflammation is mediated by the P2Y12 receptor. J Exp Med 2009; 206 (11) 2543-2555
  • 45 Cattaneo M. The platelet P2 receptors in inflammation. Hamostaseologie 2015; 35 (03) 262-266
  • 46 Amison RT, Momi S, Morris A. , et al. RhoA signaling through platelet P2Y1 receptor controls leukocyte recruitment in allergic mice. J Allergy Clin Immunol 2015; 135 (02) 528-538
  • 47 Hagiwara S, Iwasaka H, Hasegawa A. , et al. Adenosine diphosphate receptor antagonist clopidogrel sulfate attenuates LPS-induced systemic inflammation in a rat model. Shock 2011; 35 (03) 289-292
  • 48 Liverani E, Rico MC, Tsygankov AY, Kilpatrick LE, Kunapuli SP. P2Y12 receptor modulates sepsis-induced inflammation. Arterioscler Thromb Vasc Biol 2016; 36 (05) 961-971
  • 49 Léon C, Ravanat C, Freund M, Cazenave JP, Gachet C. Differential involvement of the P2Y1 and P2Y12 receptors in platelet procoagulant activity. Arterioscler Thromb Vasc Biol 2003; 23 (10) 1941-1947
  • 50 Zerr M, Hechler B, Freund M. , et al. Major contribution of the P2Y1receptor in purinergic regulation of TNFα-induced vascular inflammation. Circulation 2011; 123 (21) 2404-2413
  • 51 Hechler B, Freund M, Ravanat C, Magnenat S, Cazenave JP, Gachet C. Reduced atherosclerotic lesions in P2Y1/apolipoprotein E double-knockout mice: the contribution of non-hematopoietic-derived P2Y1 receptors. Circulation 2008; 118 (07) 754-763
  • 52 Liu R, Ma S, Lu Z, Shen H, Sun L, Wei M. The ADP antagonist MRS2179 regulates the phenotype of smooth muscle cells to limit intimal hyperplasia. Cardiovasc Drugs Ther 2015; 29 (01) 23-29
  • 53 Marques-da-Silva C, Burnstock G, Ojcius DM, Coutinho-Silva R. Purinergic receptor agonists modulate phagocytosis and clearance of apoptotic cells in macrophages. Immunobiology 2011; 216 (1–2): 1-11
  • 54 Darbousset R, Delierneux C, Mezouar S. , et al. P2 × 1 expressed on polymorphonuclear neutrophils and platelets is required for thrombosis in mice. Blood 2014; 124 (16) 2575-2585
  • 55 Ilkan Z, Watson S, Watson SP, Mahaut-Smith MP. P2 × 1 receptors amplify FcγRIIa-induced Ca2+ increases and functional responses in human platelets. Thromb Haemost 2018; 118 (02) 369-380
  • 56 Lecut C, Faccinetto C, Delierneux C. , et al. ATP-gated P2 × 1 ion channels protect against endotoxemia by dampening neutrophil activation. J Thromb Haemost 2012; 10 (03) 453-465
  • 57 Maître B, Magnenat S, Heim V. , et al. The P2 × 1 receptor is required for neutrophil extravasation during lipopolysaccharide-induced lethal endotoxemia in mice. J Immunol 2015; 194 (02) 739-749
  • 58 El Mdawar MB, Maître B, Magnenat S, Gachet C, Hechler B, de la Salle H. The ATP-gated P2X1 ion channel contributes to the severity of antibody-mediated transfusion-related acute lung injury in mice. Sci Rep 2019; 9 (01) 5159
  • 59 Sesma JI, Weitzer CD, Livraghi-Butrico A. , et al. UDP-glucose promotes neutrophil recruitment in the lung. Purinergic Signal 2016; 12 (04) 627-635
  • 60 Amison RT, Arnold S, O'Shaughnessy BG. , et al. Lipopolysaccharide (LPS) induced pulmonary neutrophil recruitment and platelet activation is mediated via the P2Y1 and P2Y14 receptors in mice. Pulm Pharmacol Ther 2017; 45: 62-68

Address for correspondence

Christian Gachet, MD, PhD
Etablissement Français du Sang-Grand Est
INSERM UMR_S1255, 10, rue Spielmann, BP 36, F-67065 Strasbourg Cedex
France   

  • References

  • 1 Gaarder A, Jonsen J, Laland S, Hellem A, Owren PA. Adenosine diphosphate in red cells as a factor in the adhesiveness of human blood platelets. Nature 1961; 192: 531-532
  • 2 Born GVR. Aggregation of blood platelets by adenosine diphosphate and its reversal. Nature 1962; 194: 927-929
  • 3 Maffrand JP, Bernat A, Delebassée D, Defreyn G, Cazenave JP, Gordon JL. ADP plays a key role in thrombogenesis in rats. Thromb Haemost 1988; 59 (02) 225-230
  • 4 Cattaneo M, Canciani MT, Lecchi A. , et al. Released adenosine diphosphate stabilizes thrombin-induced human platelet aggregates. Blood 1990; 75 (05) 1081-1086
  • 5 Dovlatova N, Wijeyeratne YD, Fox SC. , et al. Detection of P2Y(14) protein in platelets and investigation of the role of P2Y(14) in platelet function in comparison with the EP(3) receptor. Thromb Haemost 2008; 100 (02) 261-270
  • 6 Hechler B, Léon C, Vial C. , et al. The P2Y1 receptor is necessary for adenosine 5′-diphosphate-induced platelet aggregation. Blood 1998; 92 (01) 152-159
  • 7 Léon C, Hechler B, Vial C, Leray C, Cazenave JP, Gachet C. The P2Y1 receptor is an ADP receptor antagonized by ATP and expressed in platelets and megakaryoblastic cells. FEBS Lett 1997; 403 (01) 26-30
  • 8 Waldo GL, Harden TK. Agonist binding and Gq-stimulating activities of the purified human P2Y1 receptor. Mol Pharmacol 2004; 65 (02) 426-436
  • 9 Crittenden JR, Bergmeier W, Zhang Y. , et al. CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation. Nat Med 2004; 10 (09) 982-986
  • 10 Gachet C. Regulation of platelet functions by P2 receptors. Annu Rev Pharmacol Toxicol 2006; 46: 277-300
  • 11 Hollopeter G, Jantzen HM, Vincent D. , et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 2001; 409 (6817): 202-207
  • 12 Zhang FL, Luo L, Gustafson E. , et al. ADP is the cognate ligand for the orphan G protein-coupled receptor SP1999. J Biol Chem 2001; 276 (11) 8608-8615
  • 13 Hechler B, Eckly A, Ohlmann P, Cazenave JP, Gachet C. The P2Y1 receptor, necessary but not sufficient to support full ADP-induced platelet aggregation, is not the target of the drug clopidogrel. Br J Haematol 1998; 103 (03) 858-866
  • 14 Trumel C, Payrastre B, Plantavid M. , et al. A key role of adenosine diphosphate in the irreversible platelet aggregation induced by the PAR1-activating peptide through the late activation of phosphoinositide 3-kinase. Blood 1999; 94 (12) 4156-4165
  • 15 Kauffenstein G, Hechler B, Cazenave JP, Gachet C. Adenine triphosphate nucleotides are antagonists at the P2Y receptor. J Thromb Haemost 2004; 2 (11) 1980-1988
  • 16 Cattaneo M. The platelet P2Y12 receptor for adenosine diphosphate: congenital and drug-induced defects. Blood 2011; 117 (07) 2102-2112
  • 17 Gremmel T, Michelson AD, Frelinger III AL, Bhatt DL. Novel aspects of antiplatelet therapy in cardiovascular disease. Res Pract Thromb Haemost 2018; 2 (03) 439-449
  • 18 Gachet C. P2Y(12) receptors in platelets and other hematopoietic and non-hematopoietic cells. Purinergic Signal 2012; 8 (03) 609-619
  • 19 Hechler B, Gachet C. Purinergic receptors in thrombosis and inflammation. Arterioscler Thromb Vasc Biol 2015; 35 (11) 2307-2315
  • 20 Hao Y, Wang L, Chen H. , et al. Targetable purinergic receptors P2Y12 and A2b antagonistically regulate bladder function. JCI Insight 2019; 4 (16) 122112
  • 21 Gratacap MP, Guillermet-Guibert J, Martin V. , et al. Regulation and roles of PI3Kβ, a major actor in platelet signaling and functions. Adv Enzyme Regul 2011; 51 (01) 106-116
  • 22 Stefanini L, Paul DS, Robledo RF. , et al. RASA3 is a critical inhibitor of RAP1-dependent platelet activation. J Clin Invest 2015; 125 (04) 1419-1432
  • 23 Lecchi A, Femia EA, Paoletta S. , et al. Inherited dysfunctional platelet P2Y12 receptor mutations associated with bleeding disorders. Hamostaseologie 2016; 36 (04) 279-283
  • 24 Hechler B, Lenain N, Marchese P. , et al. A role of the fast ATP-gated P2 × 1 cation channel in thrombosis of small arteries in vivo. J Exp Med 2003; 198 (04) 661-667
  • 25 Cattaneo M. The P2 receptors and congenital platelet function defects. Semin Thromb Hemost 2005; 31 (02) 168-173
  • 26 Oury C, Toth-Zsamboki E, Van Geet C. , et al. A natural dominant negative P2 × 1 receptor due to deletion of a single amino acid residue. J Biol Chem 2000; 275 (30) 22611-22614
  • 27 Gachet C. Antiplatelet drugs: which targets for which treatments?. J Thromb Haemost 2015; 13 (Suppl. 01) S313-S322
  • 28 Zhang J, Zhang K, Gao ZG. , et al. Agonist-bound structure of the human P2Y12 receptor. Nature 2014; 509 (7498): 119-122
  • 29 Zhang K, Zhang J, Gao ZG. , et al. Structure of the human P2Y12 receptor in complex with an antithrombotic drug. Nature 2014; 509 (7498): 115-118
  • 30 Fabre JE, Nguyen M, Latour A. , et al. Decreased platelet aggregation, increased bleeding time and resistance to thromboembolism in P2Y1-deficient mice. Nat Med 1999; 5 (10) 1199-1202
  • 31 Léon C, Hechler B, Freund M. , et al. Defective platelet aggregation and increased resistance to thrombosis in purinergic P2Y(1) receptor-null mice. J Clin Invest 1999; 104 (12) 1731-1737
  • 32 Léon C, Freund M, Ravanat C, Baurand A, Cazenave JP, Gachet C. Key role of the P2Y(1) receptor in tissue factor-induced thrombin-dependent acute thromboembolism: studies in P2Y(1)-knockout mice and mice treated with a P2Y(1) antagonist. Circulation 2001; 103 (05) 718-723
  • 33 Lenain N, Freund M, Léon C, Cazenave JP, Gachet C. Inhibition of localized thrombosis in P2Y1-deficient mice and rodents treated with MRS2179, a P2Y1 receptor antagonist. J Thromb Haemost 2003; 1 (06) 1144-1149
  • 34 Hechler B, Nonne C, Roh EJ. , et al. MRS2500 [2-iodo-N6-methyl-(N)-methanocarba-2′-deoxyadenosine-3′,5′-bisphosphate], a potent, selective, and stable antagonist of the platelet P2Y1 receptor with strong antithrombotic activity in mice. J Pharmacol Exp Ther 2006; 316 (02) 556-563
  • 35 Zhang D, Gao ZG, Zhang K. , et al. Two disparate ligand-binding sites in the human P2Y1 receptor. Nature 2015; 520 (7547): 317-321
  • 36 Morrell CN, Aggrey AA, Chapman LM, Modjeski KL. Emerging roles for platelets as immune and inflammatory cells. Blood 2014; 123 (18) 2759-2767
  • 37 Idzko M, Ferrari D, Eltzschig HK. Nucleotide signalling during inflammation. Nature 2014; 509 (7500): 310-317
  • 38 Steinhubl SR, Badimon JJ, Bhatt DL, Herbert JM, Lüscher TF. Clinical evidence for anti-inflammatory effects of antiplatelet therapy in patients with atherothrombotic disease. Vasc Med 2007; 12 (02) 113-122
  • 39 Evans DJ, Jackman LE, Chamberlain J. , et al. Platelet P2Y(12) receptor influences the vessel wall response to arterial injury and thrombosis. Circulation 2009; 119 (01) 116-122
  • 40 Li D, Wang Y, Zhang L. , et al. Roles of purinergic receptor P2Y, G protein-coupled 12 in the development of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012; 32 (08) e81-e89
  • 41 West LE, Steiner T, Judge HM, Francis SE, Storey RF. Vessel wall, not platelet, P2Y12 potentiates early atherogenesis. Cardiovasc Res 2014; 102 (03) 429-435
  • 42 Abele S, Spriewald BM, Ramsperger-Gleixner M. , et al. Attenuation of transplant arteriosclerosis with clopidogrel is associated with a reduction of infiltrating dendritic cells and macrophages in murine aortic allografts. Transplantation 2009; 87 (02) 207-216
  • 43 Harada K, Matsumoto Y, Umemura K. Adenosine diphosphate receptor P2Y12-mediated migration of host smooth muscle-like cells and leukocytes in the development of transplant arteriosclerosis. Transplantation 2011; 92 (02) 148-154
  • 44 Paruchuri S, Tashimo H, Feng C. , et al. Leukotriene E4-induced pulmonary inflammation is mediated by the P2Y12 receptor. J Exp Med 2009; 206 (11) 2543-2555
  • 45 Cattaneo M. The platelet P2 receptors in inflammation. Hamostaseologie 2015; 35 (03) 262-266
  • 46 Amison RT, Momi S, Morris A. , et al. RhoA signaling through platelet P2Y1 receptor controls leukocyte recruitment in allergic mice. J Allergy Clin Immunol 2015; 135 (02) 528-538
  • 47 Hagiwara S, Iwasaka H, Hasegawa A. , et al. Adenosine diphosphate receptor antagonist clopidogrel sulfate attenuates LPS-induced systemic inflammation in a rat model. Shock 2011; 35 (03) 289-292
  • 48 Liverani E, Rico MC, Tsygankov AY, Kilpatrick LE, Kunapuli SP. P2Y12 receptor modulates sepsis-induced inflammation. Arterioscler Thromb Vasc Biol 2016; 36 (05) 961-971
  • 49 Léon C, Ravanat C, Freund M, Cazenave JP, Gachet C. Differential involvement of the P2Y1 and P2Y12 receptors in platelet procoagulant activity. Arterioscler Thromb Vasc Biol 2003; 23 (10) 1941-1947
  • 50 Zerr M, Hechler B, Freund M. , et al. Major contribution of the P2Y1receptor in purinergic regulation of TNFα-induced vascular inflammation. Circulation 2011; 123 (21) 2404-2413
  • 51 Hechler B, Freund M, Ravanat C, Magnenat S, Cazenave JP, Gachet C. Reduced atherosclerotic lesions in P2Y1/apolipoprotein E double-knockout mice: the contribution of non-hematopoietic-derived P2Y1 receptors. Circulation 2008; 118 (07) 754-763
  • 52 Liu R, Ma S, Lu Z, Shen H, Sun L, Wei M. The ADP antagonist MRS2179 regulates the phenotype of smooth muscle cells to limit intimal hyperplasia. Cardiovasc Drugs Ther 2015; 29 (01) 23-29
  • 53 Marques-da-Silva C, Burnstock G, Ojcius DM, Coutinho-Silva R. Purinergic receptor agonists modulate phagocytosis and clearance of apoptotic cells in macrophages. Immunobiology 2011; 216 (1–2): 1-11
  • 54 Darbousset R, Delierneux C, Mezouar S. , et al. P2 × 1 expressed on polymorphonuclear neutrophils and platelets is required for thrombosis in mice. Blood 2014; 124 (16) 2575-2585
  • 55 Ilkan Z, Watson S, Watson SP, Mahaut-Smith MP. P2 × 1 receptors amplify FcγRIIa-induced Ca2+ increases and functional responses in human platelets. Thromb Haemost 2018; 118 (02) 369-380
  • 56 Lecut C, Faccinetto C, Delierneux C. , et al. ATP-gated P2 × 1 ion channels protect against endotoxemia by dampening neutrophil activation. J Thromb Haemost 2012; 10 (03) 453-465
  • 57 Maître B, Magnenat S, Heim V. , et al. The P2 × 1 receptor is required for neutrophil extravasation during lipopolysaccharide-induced lethal endotoxemia in mice. J Immunol 2015; 194 (02) 739-749
  • 58 El Mdawar MB, Maître B, Magnenat S, Gachet C, Hechler B, de la Salle H. The ATP-gated P2X1 ion channel contributes to the severity of antibody-mediated transfusion-related acute lung injury in mice. Sci Rep 2019; 9 (01) 5159
  • 59 Sesma JI, Weitzer CD, Livraghi-Butrico A. , et al. UDP-glucose promotes neutrophil recruitment in the lung. Purinergic Signal 2016; 12 (04) 627-635
  • 60 Amison RT, Arnold S, O'Shaughnessy BG. , et al. Lipopolysaccharide (LPS) induced pulmonary neutrophil recruitment and platelet activation is mediated via the P2Y1 and P2Y14 receptors in mice. Pulm Pharmacol Ther 2017; 45: 62-68