| Home | E-Submission | Sitemap | Contact Us |  
top_img
Clin Exp Thromb Hemost > Volume 9(2); 2024 > Article
Chaudhary, Kim, and Kim: Platelet Activation Through G-protein Coupled Receptors

Abstract

Platelet contains G-protein coupled receptors (GPCRs) that play a key role in platelet activation and thrombus formation in response to various agonists including ADP, thromboxane A2, thrombin, epinephrine, and serotonin by coupling to various G-proteins. Since platelets play an important role in health and diseases, platelet GPCRs have been one of the primary drug targets. However, progress should be made in our understanding of GPCR dynamics, activation, and signaling to open new possibilities for selective drug development. In this review, we have summarized the platelet GPCRs, their associated G-protein signaling, and the downstream signaling mechanism that results in both activation and desensitization of platelets. A deeper understanding of the processes behind GPCR-mediated signaling might enable antiplatelet medication to be customized in the future.

Origin of platelet

Platelets are derived from larger progenitor cells called megakaryocytes that undergo fragmentation at their final stage of development. Hematopoietic stem cells (HSCs), which reside adjacent to the endosteal bone surface, generate progenitor cells that migrate to the vessels in the marrow cavity. A single daughter cell exits the bone after each division to proliferate and develop into numerous potential lineages, one of which is the megakaryocyte. The mechanical extension of proplatelet extensions into the sinusoidal blood vessels of the bone marrow is preceded by an elaborate intracellular program of nuclear amplification and protein production in maturing megakaryocytes. The released proplatelets continue to mature in the vasculature, eventually releasing individual platelets from the tips of the platelets [1,2]. Platelets are essential for forming clots by the formation of a thrombus and help in hemostasis. Platelets circulate in the bloodstream in a dormant discoid state but can be activated when they are exposed to damaged blood vessel walls [3].

Structure of platelet

Although platelets circulate in the blood in a quiescent state, when activated, they undergo major structural changes mediated by actin and myosin within the cytoplasm. The platelets are transformed from a discoid form to a compact sphere with dendritic extensions that allow the platelets to adhere to each other. These dendritic extensions are formed from filopodia which generate sheet-like lamellipodia that strengthen platelet’s contact with the area of injury [4]. This process of formation of filopodia and lamellipodia causes shape changes in platelets and aids in the adhesion of platelet to platelet for thrombus formation. Adhesion and aggregation of platelets are facilitated through a variety of membrane-bound receptors [5]. Platelets are not nucleated, but they do contain RNA, ribosomes, mitochondria, and other granules that are essential in enacting platelet function [6]. Platelet secretory organelles are classified into three types: α-granules, dense granules, and lysosomes. These granules include a variety of platelet-specific and non-specific proteins that aid in platelet activity [7-10].

Physiological function of platelet

As described above, platelets have a crucial role in thrombus formation and hemostasis. Following vascular injury, substances in the exposed extracellular matrix, including collagen and von Willebrand factor, can bind to the surface receptors of platelets causing the adhesion of platelets [3]. This adherence of platelets to the injury site leads to the activation of platelets. Upon activation, the platelets undergo a conformational change and begin secreting granular contents that promote aggregation and platelet clumping [4].
The signaling mechanism of platelet activation is complex and is mediated mainly via GPCRs and non-GPCRs. GPCRs are seven-transmembrane receptors and are the most numerous protein family encoded by the human genome that govern practically all physiological functions through G-protein signaling in various cells including platelets [11-15]. Non-GPCRs are essential receptors that activate platelets via the Glycoprotein VI (GPVI) receptors. While G protein-mediated signaling plays a minimal role in the initial adhesion of platelets to the vessel wall, mediators acting through G protein-coupled receptors are necessary for the subsequent recruitment of additional platelets into a growing platelet thrombus. In this review, we summarized the recent progress in understanding how platelet function is regulated by GPCR signaling and what are the molecular mechanisms underlying GPCR-mediated platelet activation.

GPCR-mediated platelet activation

GPCRs are seven-transmembrane receptors located on the cell membrane and are responsible for translating extracellular signals into important physiological effects [16]. Endogenous ligands of GPCRs include hormones, neurotransmitters, chemokines, and so on ranging from photons, amines, carbohydrates, lipids, and peptides to proteins [17].
GPCR-mediated platelet activation involves three primary G-protein-mediated pathways: Gq, G12/13, and Gi. Heterotrimeric G-proteins are known to exist in their inactive state as stable complexes of Gα subunits and Gβγ dimers. G subunits cycle from a guanosine triphosphate (GDP)-bound inactive state to a GTP-bound active state, with guanosine diphosphate (GTP) hydrolysis limiting the active state’s lifespan. In vitro biochemical investigations have revealed that active G-protein heterotrimers break down into Gα-GTP and Gβ/γ subunits [18]. The most soluble agonists produced by active cells, such as ADP, thromboxane A2 (TxA2), and thrombin activate platelets via GPCRs in the coagulation cascade [19-22]. Similarly, epinephrine and serotonin are involved in activating platelets through the GPCR pathway [23].

Platelet activation by adenosine diphosphate (ADP)

At the site of vascular injury, ADP is released from the cells that act as an autocrine and paracrine stimulation for the recruitment of extra platelets and stabilize the hemostatic plug [11]. In platelet, ADP is stored in the dense granules and is released upon platelet activation. Two different GPCRs for ADP designated P2Y1 and P2Y12, are expressed in both human and mouse platelets (Fig. 1) [24,25]. P2Y1 and P2Y12 receptors couple to Gq and Gi family members of G-protein, respectively [26-29]. For complete platelet activation, co-activation of P2Y1 and P2Y12 receptors is essential [30-33]. ADP-induced platelet aggregation is impaired and there is no shape change in mice lacking P2Y1 receptors [34,35]. Additionally, mice lacking P2Y1 have a slightly prolonged bleeding time and are also relatively resistant to ADP-induced thromboembolism [36,37]. Interestingly, mice platelet deficient in P2Y12 have a normal shape change but impaired platelet aggregation in response to ADP [38,39]. In humans, deficiency of P2Y12 has shown moderate hemorrhagia, however, mice with P2Y12 deficiency do not have this trait [34,35,40,41]. Interestingly, platelet responses to low and intermediate concentrations of TxA2 and thrombin were reduced in the absence of ADP, highlighting the importance of ADP as a positive feedback mediator required for sustained platelet activation [36-38,42]. Thienopyridines such as clopidogrel, currently used in the secondary prevention of cardiovascular events, irreversibly inhibit the P2Y12 receptor [43,44].

Platelet activation by thrombin

Thrombin being one of the most potent activators of platelets, is the major effector protease of the coagulation system. After the disruption of the vascular endothelium, tissue factor is exposed to plasma coagulation factors, which causes thrombin production. Thrombin is formed on cellular surfaces, particularly those of activated platelets [45]. An important mechanism by which activated platelets stimulate coagulation is the local production of thrombin on the platelet surface. Thrombin induces platelet activation through the protease-activated receptor (PAR) family member of GPCRs (Fig. 1). Among the four members of the PAR family, three (PAR1, PAR3, and PAR4) are activated by thrombin. In human platelet, PAR1 and PAR4, and in mouse platelet PAR3 and PAR4 are expressed [46-48]. Synthetic peptides based on the sequence of the PAR1- and PAR4- tethered ligand domains can activate the receptors, imitating at least part of the activities of thrombin. PAR3 receptor in human platelet signal in response to the thrombin in transfected cells while in murine platelets it is mainly involved in facilitating PAR4 receptor cleavage rather than in signaling by its own [49]. Platelets can be activated by thrombin at concentrations as low as 0.1 nM. While various platelet agonists can trigger hydrolysis of phosphoinositide, none appear to couple to phospholipase C (PLC), an important enzyme that hydrolyzes the membrane-bound phosphatidylinositol 4,5-bisphosphate (PIP2) to produce secondary messenger as efficiently as thrombin [50]. PARs that couple to the Gq and G12/G13 family of heterotrimeric G proteins which are directly involved in the thrombin-induced platelet activation (Fig. 1) [51-53]. PAR1 antagonists or antibodies that prevent PAR1 or PAR4 activation have been used in studies to show that PAR1 causes human platelet activation at low thrombin concentrations, but PAR4 only contributes to thrombin-induced platelet activation at high thrombin concentrations [46,54-56]. It is assumed that the presence of PAR1 and PAR4 in human platelets is analogous to the PAR3 and PAR4 in murine platelets which is indicated by the study using PAR3-deficient mice which did not respond to any low and intermediate concentrations of thrombin but could be activated by high concentrations via PAR4 [46]. The greater potency of thrombin to activate PAR1 is most likely due to the presence of a hirudin-like sequence adjacent to the C-terminal thrombin cleavage site, which enhances thrombin binding and is missing in PAR4 [46,57]. However, experiments using heterologously produced receptors indicated that unless PAR4 was present, mouse PAR3, which has a hirudin-like region near the thrombin cleavage site, did not mediate thrombin-induced transmembrane signaling. This shows that at low thrombin concentrations, mouse PAR3 acts as a co-receptor for mouse PAR4 and enhances cleavage and activation of PAR4 [48]. Similarly, PAR4 knockout mice’s platelets are unresponsive to thrombin and are protected from thrombosis [58,59]. Thus, thrombin-induced platelet activation in mice is completely dependent on PAR4-mediated signaling and requires only PAR3 to facilitate PAR4 cleavage at low thrombin concentrations, whereas thrombin activates human platelets by cleaving and activating PAR1 and PAR4 at low and high concentrations, respectively. It is not clear that in mouse PAR3 and PAR4 forms stable heterodimers in which the former promotes the activation and the latter helps in signaling or whether thrombin binding to PAR3 only aids PAR4 cleavage [11].

Platelet activation by TxA2

TxA2 release, similar to ADP secretion, enhances the initial stimulus for the activation of platelets and aids in the recruitment of more platelets. The short half-life of TxA2 in solution is the limiting factor of this process which helps to restrict the spread of platelet activation to the initial site of injury. The activities of the TxA2 are restricted to the local area only because of its half-life. TxA2 is synthesized from arachidonic acid where cyclooxygenase-1 enzyme is used which targets the low-dose aspirin and thromboxane synthase. Thromboxane exerts its effect via the TxA2 receptor (TP). This receptor is also known to activate through prostaglandin endoperoxides PGG2 and PGH2. TP receptor couples to the Gq and G12/G13 family protein of GPCR and exerts downstream signaling in platelet [52,60,61]. Studies using platelets from TP-deficient mice suggested that platelets do not respond to TxA2, which indicate the role of TP as a TxA2 receptor on platelets [62]. Additionally, TP-knockout mice show increased bleeding time and are not able to form stable thrombus [62]. It has been suggested that reduced activation of TP-deficient platelets may contribute to a reduction in injury-induced vascular proliferation and atherosclerotic progression [63,64].

Platelet activation by epinephrine

Epinephrine, also called adrenaline is both a hormone and a neurotransmitter. In human platelets, epinephrine is a weak activator, compared to thrombin. However, in the case of humans, mild bleeding disorder is related to reduced epinephrine-induced aggregation. Epinephrine has the ability to enhance the effects of other agonists in both murine and human platelets. Epinephrine induces platelet aggregation mediated through α2A-adrenergic receptors [23,65,66]. It is suggested that epinephrine induces platelet activation through its receptor coupled to Gi family members of GPCR (Fig. 1). Epinephrine couples to Gz which belongs to the Gi class of G-protein [67]. Knockout studies show that loss of G expression abolishes adrenergic responses in mouse platelets, whereas loss of Gi does not. The study revealed that epinephrine responses in mouse platelets are nullified when G expression is abolished, whereas Gi-knockout has no effect. The ability of adrenaline to activate Rap1b also appears to be mediated by Gz [68,69].

Platelet activation by serotonin

Platelets take up serotonin and are stored in dense granules. They are only released after activation of the platelet. The concentration of serotonin is higher in dense granules, however, platelets are unable to produce serotonin due to the lack of an enzyme (5-hydroxytryptophan decarboxylase) to cause decarboxylation [70,71]. Serotonin is able to induce its responses through the platelet surface 5-HT2A receptor which is a member of GPCR (Fig. 1) [72-75]. Serotonin binding to 5-HT2A initiates typical Gq-linked signal transduction events via the PLC pathway, promoting the production of inositol triphosphate (IP3) and diacylglycerol (DAG), culminating in calcium mobilization [76-78].
Interestingly, it has been found that serotonin alone only induces shape change while epinephrine alone induces neither shape change nor aggregation in platelets [79]. It has been known that the co-stimulation of epinephrine and serotonin induces platelet aggregation by bypassing the P2Y1 and P2Y12 receptors and activates Gq-coupled 5HT2A and Gz coupled α2A adrenergic receptors respectively [80].

G protein–mediated platelet signaling pathways

ADP, TxA2, and thrombin are the principal diffusible platelet stimuli that attract platelets into a developing thrombus by activating numerous G protein-mediated signaling pathways that produce platelet-shape change, degranulation, and integrin αIIβ3-mediated aggregation. For the complete activation of platelets, agonists use different mechanisms mediated via G-protein. As described above, ADP induces platelet activation binding to P2Y1 and P2Y12 receptors and activates Gq and Gi respectively. Similarly, thromboxane binds to the TP receptor and thrombin binds to protease-activated receptors PAR1/PAR4 or PAR3/PAR4 which activates G12/G13 and Gq [24,25,46,47]. All these mediators increase the formation and release of ADP, TxA2, and thrombin which ultimately amplifies the G-protein-mediated signaling pathway. This positive feedback mechanism is limiting the analysis of the role of individual G-protein-mediated signaling pathways involved in platelet activation. Studies with α subunits knockout platelets to analyze the G-protein mediated signaling pathway independent of agonist use and their receptors have shown that the G proteins Gq, G12/13, and Gi2 (Table 1) play a crucial role in platelet activation [11].

Involvement of Gq protein in platelet signaling

Upon agonist stimulation, PLCβ is produced by the Gq family of GPCR. This family protein is essential for the granular secretion of platelets and integrin activation which results in platelet aggregation [81]. The downstream signaling of the Gq initiates with PLCβ activation which in turn leads to the formation of IP3 and DAG, which increase cytosolic free calcium [Ca2+] and activate protein kinase C (PKC) [81,82]. In addition, there is also involvement of βγ-complex which interacts with phosphatidylinositol 3-kinase (PI3K), a downstream effector to induce platelet activation (Fig. 1) [83]. Although the majority of the mammalian cells express both Gq and G11, interestingly platelet is only the exception which contains only Gq [81,82]. To date there is no physiological significance has been reported for the absence of G11 in blood platelets. On platelet activation with several agonists including ADP, thrombin, and TxA2 analog U46619 in Gq deficient mice, there is a decrease in granular secretion and aggregation [81]. Furthermore, in ADP-triggered shape change in platelets, Gq plays a crucial role most likely by stimulating calcium/calmodulin-dependent and/or RhoA-dependent signaling mechanism [84]. Platelet activation requires an increase in intracellular calcium. Increased calcium has a variety of consequences, including platelet shape alteration and activation of small GTPases, which increase platelet granule exocytosis [85,86]. It is worth noting that, in comparison to the degree of calcium mobilization elicited by ADP or thrombin activation, serotonin through the 5-HT2A results in a lesser calcium influx [26,30]. This is in line with the research that describes serotonin as a helper agonist that stimulates platelet aggregation along with others and cannot induce sufficient platelet aggregation on its own but can potentiate the aggregation combined with other stimuli [26,87].

Platelet signaling induced by involvement of G12/13 protein

The G12/G13 protein family is abundantly expressed in platelets which show the Rho/Rho-kinase–mediated signaling pathway in platelet activation [52]. G12/13 is bound to GTP which interacts and activates guanine nucleotide exchange factors (GEFs) for the p115Rho-GEF protein. p115RhoGEF protein helps to convert GDP-bound RhoA to GTP-bound RhoA [88-90]. Similarly, Rho kinases are phosphorylated after activation by RhoA and inhibit myosin light chain phosphatase [53]. As a result, phosphorylation of the myosin light chain as well as contraction via myosin light chain are enhanced. Therefore, G12/13 promotes shape change in platelets along with granular secretion [53]. However, platelet aggregation in response to low doses of thrombin and TxA2 analog U46619 in G13-knockout platelets is reduced whereas granular secretion is reduced in response to U46619 but not when induced by thrombin [91]. G13-knockout platelets show a reduction in the shape change when induced by the agonists. Interestingly, G12/13 plays a critical role in integrin outside-in signaling by binding to the cytoplasmic domain of integrin [92].

Platelet signaling induced by involvement of Gi protein

In the Gi family protein, Gi2 is abundantly expressed in platelets, upon agonist couple to the receptor, the Gi2-α subunit inhibits the adenylyl cyclase enzyme and terminates the further downstream signaling [93]. Furthermore, Gi-βγ complexes, which are generated upon G-protein activation and have the ability to control a range of channels or enzymes such as PI3Ks and adenylyl cyclases [93]. The enzyme PI3Ks triggers a number of downstream effectors such as AKT/protein kinase B (PKB), serine/threonine kinase, ERK, Rap1b, and Src family kinases (Fig. 1). In platelet, ADP-activated receptor P2Y12-coupled to Gi protein plays a key role in PI3K activation, specifically, βγ-subunit activated PI3K, which blocks RASA3 and causes persistently active Rap1b [94-96]. Activation of P2Y12 results in prolonged active Rap1b because it inhibits adenylate cyclase, which lowers cAMP, and activates PI3K, which inhibits RASA3 [97].

Orphan G protein-coupled receptors

Orphan GPCRs are receptors that lack endogenous ligands [98]. Currently, there are more than 140 GPCRs that have unidentified endogenous ligands which create doubt in the actual function of discovered GPCRs. These orphan GPCRs can have great potential for therapeutic targets [99].
Recently, GPR31 has been known to show the signaling through the Gi protein family which ultimately enhances the PAR-4 mediated platelet activation in human and mouse. Additionally, it has been known that the involvement of GPR31 also results in Rap1 and p38 activation and little calcium flux, however, it fails to induce platelet aggregation [100]. Similarly, GPR56 is also known to cause activation of G13 signaling leading to platelet shape change. Additionally, in GPR56 knockout mice it has been found that there is prolonged bleeding and impaired platelet plug formation [101]. GPR55, another orphan receptor, is also known to induce platelet aggregation in humans but the mechanism is still unknown [102-104]. Further research is essential to fully understand how these orphan receptors affect platelet signaling and whether the knowledge can be translated from bench to clinics.

GPCR signaling in platelet activation: recent developments

In recent years, it has been shown that platelets are not only activated but also desensitized through GPCRs. Several mechanisms prevent the hyperactivation of GPCR signaling in various cells, among which G Protein-Coupled Receptor Kinases (GRKs) and arrestins play an important role and these are therefore gaining popularity among researchers [105,106]. When GPCR is activated by ligand binding, G protein coupling, and signal transduction occur, GRK enters the picture and phosphorylates the active GPCR’s cytoplasmic serine and threonine residues, opening the door for arrestin recruitment. Desensitization is the process by which the recruited arrestin attaches to the phosphorylated GPCR, prevents further G-protein coupling, and stops the downstream signaling [107-110]. There are 7 isoforms for GRKs among which GRK2, GRK3 GRK5, and GRK6, and 4 isoforms of β-arrestin among which β-arrestin 1 and β-arrestin 2 are known to present abundantly in platelet [79,111-116]. There is a dearth of knowledge on the regulation and mechanisms of GPCR signaling and GPCR desensitization by GRKs in platelets, despite the crucial roles played by these cells in GPCR activities. We and others have just lately shown the intriguing aspects of the arrestin and GRK system in platelet GPCR signaling. We demonstrated using GRK6 knockout mouse platelets that GPCR agonists, such as 2-MeSADP, U46619 (TxA2 analog), AYPGKF, and thrombin, dramatically increase platelet aggregation and dense granule production in comparison to wildtype platelets [79]. GRK6-deficient platelets do not, however, exhibit GPVI agonist collagen-related peptide (CRP)-induced platelet aggregation or dense granule secretion. This suggests that GRK6 regulates P2Y1, P2Y12, TPα, and PARs-mediated signaling in platelets, but not non-GPCR-mediated platelet activation. Recent research revealed that GRK6 contributes to ADP-induced P2Y12 receptor desensitization in platelets, but not P2Y1 receptor desensitization [115]. While co-stimulation of serotonin and epinephrine mimics the ADP-induced P2Y1 and P2Y12 receptor-mediated platelet activation pathway, it is noteworthy that GRK6 is not involved in serotonin-induced Gq-coupled 5-HT2A receptor and epinephrine-induced Gz-coupled α2A adrenergic receptor-mediated platelet activation, in contrast to ADP-induced aggregation. This suggests that the kinase activity of GRKs varies with different ligands, receptors, and proteins. According to reports, GRK5 is involved in the desensitization of GPCR receptors and controls thrombin signaling in platelets via the PAR-1 receptor [117,118]. Similarly, we recently assessed the functional significance of arrestin isoforms and their molecular basis of modulation of GPCR desensitization in platelets using β-arrestin1 and β-arrestin2 -/- mice. Our findings demonstrated that β-arrestin2 -/- but not β-arrestin1 -/- potentiates the aggregation and secretion of platelets triggered by agonists, including thrombin, ADP, AYPGKF, and U46619. In platelets lacking β-arrestin2, co-stimulation of serotonin and adrenaline enhanced platelet aggregation, indicating that β-arrestin2 is crucial for controlling overall GPCR signaling. Furthermore, we demonstrated that β-arrestin2 controls the desensitization of ADP and PAR4 receptors, which in turn controls the Gq- and Gi-mediated signaling in platelets. β-arrestin2 was additionally involved in thrombus formation in vivo. It is yet unclear, nevertheless, whether or not platelet GPCRs are phosphorylated by GRK isoforms other than GRK6 and GRK5, and whether or not this phosphorylation alters the receptor’s function. Furthermore, whereas arrestins play a crucial role in GPCR-mediated signaling, the process by which arrestins desensitize GPCRs in platelets remains unclear. We can find a new way to develop drugs in the field of vascular pathobiology by evaluating the functional significance of individual GRK and β-arrestin isoforms in platelets and characterizing the novel mechanisms involved in GPCR desensitization and trafficking in platelets using knockout mice for each isoform.

Conclusion and recommending the way forward

GPCRs are the largest and most diverse class of signaling proteins that affect an incredible array of physiological processes throughout the human body. Platelets operate mainly through the GPCR-linked G-protein signaling mechanisms after the stimulation with different agonists. Based on the diverse platelet physical, biochemical, and functional variability, the dynamic nature of platelet properties in health and disease varies. Nevertheless, there are significant gaps in our understanding of platelet function in these other disorders that prohibit us from reaching a thorough mechanistic understanding. Studying platelets and platelet GPCRs can provide insights into identifying several GPCR-targeted therapies in platelet-associated diseases in the future.

Acknowledgments

This work was supported by a grant from the Korean Society on Thrombosis and Hemostasis (KSTH 2021- 000).

Fig. 1.
Platelet signaling pathway through the GPCRs. Downstream signaling mechanisms linking to GPCR activation by different agonists involving different G-proteins. PLC-β, PhospholipaseC-β; PIP2, phosphatidylinositol 4,5 -bisphosphate; PIP3, phosphatidylinositol-3,4,5-trisphosphate; RhoGEF indicates Rho-guanine nucleotide exchange factor; MLCK, Myosin light chain kinase; DAG, diacyl glycerol; CalDAG-GEF, calcium and diacyl glycerol-regulated guanine nucleotide exchange factor; RIAM, Rap1–GTP-interacting adapter molecule; PKC, Protein Kinase C; RhoA, Ras homolog family member A; AC, adenylate cyclase; cAMP, 3’,5’-cyclic adenosine monophosphate; MAPK, Mitogen-activated protein kinase kinase.
ceth-9-2-20f1.jpg
Table 1.
Various GPCRs and G-proteins involved in platelet signaling
GPCR agonist GPCR receptor G protein α and β/γ subunit Effector Platelet phenotype References
1 ADP P2Y12 i AC↓ ➢ Agonist-induced inhibition of cAMP formation and reduces aggregation. [29, 69]
2 Epinephrine α2A-adenergic receptor ➢ Shape change is normal.
3 ADP P2Y1 q PLC-β ↑ ➢ Enhance IP3 production; cytosolic Ca2+ production, aggregation and secretion. [69, 81, 119]
4 Thromboxane TP
5 Thrombin PAR ➢ Shape change is normal.
6 Serotonin 5HT2A ➢ Activation of RhoA protein.
7 Thromboxane TP 12/13 RhoGEF↑ ➢ Enhance aggregation and secretion. [93]
8 Thrombin PAR ➢ Shape change is impaired.
➢ RhoA activation is enhanced.
9 ADP P2Y12 Gβ/γ PI3Kβ/γ↑ ➢ Activates PI3K and AKT, Rap1, ERK, Src family kinases. [29, 69, 120]
➢ Normal shape change.
➢ Increase cytoplasmic calcium release, granular secretion.

References

1. Italiano JE Jr, Lecine P, Shivdasani RA, Hartwig JH. Blood platelets are assembled principally at the ends of proplatelet processes produced by differentiated megakaryocytes. J Cell Biol 1999;147:1299-312.
crossref pmid pmc
2. Yin T, Li L. The stem cell niches in bone. J Clin Invest 2006;116:1195-201.
crossref pmid pmc
3. Holinstat M. Normal platelet function. Cancer Metastasis Rev 2017;36:195-8.
crossref pmid pmc
4. Aslan JE, Itakura A, Gertz JM, McCarty OJ. Platelet shape change and spreading. Methods Mol Biol 2012;788:91-100.
crossref pmid
5. George JN. Platelets. Lancet 2000;355:1531-9.
crossref pmid
6. Montague SJ, Lim YJ, Lee WM, Gardiner EE. Imaging platelet processes and function-current and emerging approaches for imaging in vitro and in vivo. Front Immunol 2020;11:78.
crossref pmid pmc
7. Sixma JJ, Slot JW, Geuze HJ. Immunocytochemical localization of platelet granule proteins. Methods Enzymol 1989;169:301-11.
pmid
8. Harrison P, Savidge GF, Cramer EM. The origin and physiological relevance of alpha-granule adhesive proteins. Br J Haematol 1990;74:125-30.
crossref pmid
9. Holmsen H, Weiss HJ. Secretable storage pools in platelets. Annu Rev Med 1979;30:119-34.
crossref pmid
10. Israels SJ, McMillan EM, Robertson C, Singhory S, McNicol A. The lysosomal granule membrane protein, LAMP-2, is also present in platelet dense granule membranes. Thromb Haemost 1996;75:623-9.
crossref pmid
11. Offermanns S. Activation of platelet function through G protein-coupled receptors. Circ Res 2006;99:1293-304.
crossref pmid
12. Vassilatis DK, Hohmann JG, Zeng H, Li F, Ranchalis JE, Mortrud MT, et al. The G protein-coupled receptor repertoires of human and mouse. Proc Natl Acad Sci USA 2003;100:4903-8.
crossref pmid pmc
13. Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmembrane receptors. Nat Rev Mol Cell Biol 2002;3:639-50.
crossref pmid
14. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB. The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol 2003;63:1256-72.
crossref pmid
15. Bockaert J, Claeysen S, Becamel C, Pinloche S, Dumuis A. G protein-coupled receptors: dominant players in cell-cell communication. Int Rev Cytol 2002;212:63-132.
crossref pmid
16. Insel PA, Sriram K, Gorr MW, Wiley SZ, Michkov A, Salmeron C, et al. An approach to discover gpcr drug targets. Trends Pharmacol Sci 2019;40:378-87.
pmid pmc
17. Sriram K, Insel PA. G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs. Mol Pharmacol 2018;93:251-8.
crossref pmid pmc
18. Gilman AG. G proteins: transducers of receptor-generated signals. Annu Rev Biochem 1987;56:615-49.
crossref pmid
19. Ghoshal K, Bhattacharyya M. Overview of platelet physiology: its hemostatic and nonhemostatic role in disease pathogenesis. Sci World J 2014;2014:781857.
crossref pmid pmc
20. Thomas MR, Storey RF. The role of platelets in inflammation. Thromb Haemost 2015;114:449-58.
crossref pmid
21. Herter JM, Rossaint J, Zarbock A. Platelets in inflammation and immunity. J Thromb Haemost 2014;12:1764-75.
crossref pmid
22. Stegner D, Nieswandt B. Platelet receptor signaling in thrombus formation. J Mol Med (Berl) 2011;89:109-21.
crossref pmid
23. Motulsky HJ, Insel PA. [3H]Dihydroergocryptine binding to alpha-adrenergic receptors of human platelets. A reassessment using the selective radioligands [3H]prazosin, [3H]yohimbine, and [3H]rauwolscine. Biochem Pharmacol 1982;31:2591-7.
pmid
24. Murugappa S, Kunapuli SP. The role of ADP receptors in platelet function. Front Biosci 2006;11:1977-86.
crossref pmid
25. Gachet C. Regulation of platelet functions by P2 receptors. Annu Rev Pharmacol Toxicol 2006;46:277-300.
crossref pmid
26. Savi P, Beauverger P, Labouret C, Delfaud M, Salel V, Kaghad M, et al. Role of P2Y1 purinoceptor in ADP-induced platelet activation. FEBS Lett 1998;422:291-5.
crossref pmid
27. Hechler B, Leon C, Vial C, Vigne P, Frelin C, Cazenave JP, et al. The P2Y1 receptor is necessary for adenosine 5’-diphosphate-induced platelet aggregation. Blood 1998;92:152-9.
crossref pmid
28. Ohlmann P, Laugwitz KL, Nurnberg B, Spicher K, Schultz G, Cazenave JP, et al. The human platelet ADP receptor activates Gi2 proteins. Biochem J 1995;312:775-9.
crossref pmid pmc
29. Jantzen HM, Milstone DS, Gousset L, Conley PB, Mortensen RM. Impaired activation of murine platelets lacking G alpha(i2). J Clin Invest 2001;108:477-83.
crossref pmid pmc
30. Jin J, Kunapuli SP. Coactivation of two different G protein-coupled receptors is essential for ADP-induced platelet aggregation. Proc Natl Acad Sci USA 1998;95:8070-4.
crossref
31. Pulcinelli FM, Pesciotti M, Pignatelli P, Riondino S, Gazzaniga PP. Concomitant activation of Gi and Gq protein-coupled receptors does not require an increase in cytosolic calcium for platelet aggregation. FEBS Lett 1998;435:115-8.
crossref
32. Jantzen HM, Gousset L, Bhaskar V, Vincent D, Tai A, Reynolds EE, et al. Evidence for two distinct G-protein-coupled ADP receptors mediating platelet activation. Thromb Haemost 1999;81:111-7.
crossref
33. Missy K, Plantavid M, Pacaud P, Viala C, Chap H, Payrastre B. Rho-kinase is involved in the sustained phosphorylation of myosin and the irreversible platelet aggregation induced by PAR1 activating peptide. Thromb Haemost 2001;85:514-20.
crossref pmid
34. Foster CJ, Prosser DM, Agans JM, Zhai Y, Smith MD, Lachowicz JE, et al. Molecular identification and characterization of the platelet ADP receptor targeted by thienopyridine antithrombotic drugs. J Clin Invest 2001;107:1591-8.
crossref pmid pmc
35. Andre P, Prasad KS, Denis CV, He M, Papalia JM, Hynes RO, et al. CD40L stabilizes arterial thrombi by a beta3 integrin-dependent mechanism. Nat Med 2002;8:247-52.
crossref pmid
36. Leon C, Hechler B, Freund M, Eckly A, Vial C, Ohlmann P, et al. Defective platelet aggregation and increased resistance to thrombosis in purinergic P2Y(1) receptor-null mice. J Clin Invest 1999;104:1731-7.
crossref
37. Fabre JE, Nguyen M, Latour A, Keifer JA, Audoly LP, Coffman TM, et al. Decreased platelet aggregation, increased bleeding time and resistance to thromboembolism in P2Y1-deficient mice. Nat Med 1999;5:1199-202.
crossref
38. Crittenden JR, Bergmeier W, Zhang Y, Piffath CL, Liang Y, Wagner DD, et al. CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation. Nat Med 2004;10:982-6.
crossref pmid
39. Nieswandt B, Schulte V, Zywietz A, Gratacap MP, Offermanns S. Costimulation of Gi- and G12/G13-mediated signaling pathways induces integrin alpha IIbbeta 3 activation in platelets. J Biol Chem 2002;277:39493-8.
pmid
40. Hollopeter G, Jantzen HM, Vincent D, Li G, England L, Ramakrishnan V, et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 2001;409:202-7.
crossref pmid
41. Cattaneo M, Zighetti ML, Lombardi R, Martinez C, Lecchi A, Conley PB, et al. Molecular bases of defective signal transduction in the platelet P2Y12 receptor of a patient with congenital bleeding. Proc Natl Acad Sci USA 2003;100:1978-83.
crossref pmid pmc
42. Dorsam RT, Kim S, Jin J, Kunapuli SP. Coordinated signaling through both G12/13 and G(i) pathways is sufficient to activate GPIIb/IIIa in human platelets. J Biol Chem 2002;277:47588-95.
crossref pmid
43. Quinn MJ, Fitzgerald DJ. Ticlopidine and clopidogrel. Circulation 1999;100:1667-72.
crossref pmid
44. Bhatt DL, Topol EJ. Scientific and therapeutic advances in antiplatelet therapy. Nat Rev Drug Discov 2003;2:15-28.
crossref pmid
45. Heemskerk JW, Bevers EM, Lindhout T. Platelet activation and blood coagulation. Thromb Haemost 2002;88:186-93.
crossref pmid
46. Kahn ML, Zheng YW, Huang W, Bigornia V, Zeng D, Moff S, et al. A dual thrombin receptor system for platelet activation. Nature 1998;394:690-4.
crossref pmid
47. Stalker TJ, Newman DK, Ma P, Wannemacher KM, Brass LF. Platelet signaling. Handb Exp Pharmacol 2012;210:59-85.
crossref pmid pmc
48. Kahn ML, Nakanishi-Matsui M, Shapiro MJ, Ishihara H, Coughlin SR. Protease-activated receptors 1 and 4 mediate activation of human platelets by thrombin. J Clin Invest 1999;103:879-87.
crossref pmid pmc
49. Nakanishi-Matsui M, Zheng YW, Sulciner DJ, Weiss EJ, Ludeman MJ, Coughlin SR. PAR3 is a cofactor for PAR4 activation by thrombin. Nature 2000;404:609-13.
crossref pmid
50. Barr AJ, Brass LF, Manning DR. Reconstitution of receptors and GTP-binding regulatory proteins (G proteins) in Sf9 cells. A direct evaluation of selectivity in receptor. G protein coupling. J Biol Chem 1997;272:2223-9.
pmid
51. Coughlin SR. Protease-activated receptors in hemostasis, thrombosis and vascular biology. J Thromb Haemost 2005;3:1800-14.
crossref pmid
52. Offermanns S, Laugwitz KL, Spicher K, Schultz G. G proteins of the G12 family are activated via thromboxane A2 and thrombin receptors in human platelets. Proc Natl Acad Sci USA 1994;91:504-8.
crossref pmid pmc
53. Klages B, Brandt U, Simon MI, Schultz G, Offermanns S. Activation of G12/G13 results in shape change and Rho/Rho-kinase-mediated myosin light chain phosphorylation in mouse platelets. J Cell Biol 1999;144:745-54.
crossref pmid pmc
54. Hung DT, Wong YH, Vu TK, Coughlin SR. The cloned platelet thrombin receptor couples to at least two distinct effectors to stimulate phosphoinositide hydrolysis and inhibit adenylyl cyclase. J Biol Chem 1992;267:20831-4.
crossref pmid
55. Bernatowicz MS, Klimas CE, Hartl KS, Peluso M, Allegretto NJ, Seiler SM. Development of potent thrombin receptor antagonist peptides. J Med Chem 1996;39:4879-87.
crossref pmid
56. Brass LF, Vassallo RR Jr, Belmonte E, Ahuja M, Cichowski K, Hoxie JA. Structure and function of the human platelet thrombin receptor. Studies using monoclonal antibodies directed against a defined domain within the receptor N terminus. J Biol Chem 1992;267:13795-8.
crossref pmid
57. Xu WF, Andersen H, Whitmore TE, Presnell SR, Yee DP, Ching A, et al. Cloning and characterization of human protease-activated receptor 4. Proc Natl Acad Sci USA 1998;95:6642-6.
crossref
58. Sambrano GR, Weiss EJ, Zheng YW, Huang W, Coughlin SR. Role of thrombin signaling in platelets in haemostasis and thrombosis. Nature 2001;413:74-8.
crossref pmid
59. Hamilton JR, Cornelissen I, Coughlin SR. Impaired hemostasis and protection against thrombosis in protease-activated receptor 4-deficient mice is due to lack of thrombin signaling in platelets. J Thromb Haemost 2004;2:1429-35.
crossref pmid
60. Knezevic I, Borg C, Le Breton GC. Identification of Gq as one of the G-proteins which copurify with human platelet thromboxane A2/prostaglandin H2 receptors. J Biol Chem 1993;268:26011-7.
crossref pmid
61. Djellas Y, Manganello JM, Antonakis K, Le Breton GC. Identification of Galpha13 as one of the G-proteins that couple to human platelet thromboxane A2 receptors. J Biol Chem 1999;274:14325-30.
pmid
62. Thomas DW, Mannon RB, Mannon PJ, Latour A, Oliver JA, Hoffman M, et al. Coagulation defects and altered hemodynamic responses in mice lacking receptors for thromboxane A2. J Clin Invest 1998;102:1994-2001.
crossref pmid pmc
63. Cheng Y, Austin SC, Rocca B, Koller BH, Coffman TM, Grosser T, et al. Role of prostacyclin in the cardiovascular response to thromboxane A2. Science 2002;296:539-41.
crossref pmid
64. Kobayashi T, Tahara Y, Matsumoto M, Iguchi M, Sano H, Murayama T, et al. Roles of thromboxane A(2) and prostacyclin in the development of atherosclerosis in apoE-deficient mice. J Clin Invest 2004;114:784-94.
crossref pmid pmc
65. Kaywin P, McDonough M, Insel PA, Shattil SJ. Platelet function in essential thrombocythemia. Decreased epinephrine responsiveness associated with a deficiency of platelet alpha-adrenergic receptors. N Engl J Med 1978;299:505-9.
crossref pmid
66. Newman KD, Williams LT, Bishopric NH, Lefkowitz RJ. Identification of alpha-adrenergic receptors in human platelets by [3H] dihydroergocryptine binding. J Clin Invest 1978;61:395-402.
crossref pmid pmc
67. Cerrito F, Lazzaro MP, Gaudio E, Arminio P, Aloisi G. 5HT2-receptors and serotonin release: their role in human platelet aggregation. Life Sci 1993;53:209-15.
crossref pmid
68. Woulfe D, Jiang H, Mortensen R, Yang J, Brass LF. Activation of Rap1B by G(i) family members in platelets. J Biol Chem 2002;277:23382-90.
crossref pmid
69. Yang J, Wu J, Jiang H, Mortensen R, Austin S, Manning DR, et al. Signaling through Gi family members in platelets. Redundancy and specificity in the regulation of adenylyl cyclase and other effectors. J Biol Chem 2002;277:46035-42.
pmid
70. Ziu E, Mercado CP, Li Y, Singh P, Ahmed BA, Freyaldenhoven S, et al. Down-regulation of the serotonin transporter in hyperreactive platelets counteracts the pro-thrombotic effect of serotonin. J Mol Cell Cardiol 2012;52:1112-21.
crossref pmid pmc
71. Gaddum JH, Giarman NJ. Preliminary studies on the biosynthesis of 5-hydroxytryptamine. Br J Pharmacol Chemother 1956;11:88-92.
crossref pmid pmc
72. Lopez-Vilchez I, Diaz-Ricart M, White JG, Escolar G, Galan AM. Serotonin enhances platelet procoagulant properties and their activation induced during platelet tissue factor uptake. Cardiovasc Res 2009;84:309-16.
crossref pmid
73. Oliver KH, Duvernay MT, Hamm HE, Carneiro AM. Loss of serotonin transporter function alters adp-mediated glycoprotein alphaiibbeta3 activation through dysregulation of the 5-HT2A receptor. J Biol Chem 2016;291:20210-9.
pmid pmc
74. Popova NK, Naumenko VS. [Polymorphism in 5-HT receptors as the background of serotonin functional diversity]. Ross Fiziol Zh Im I M Sechenova 2010;96:778-86.
pmid
75. Barnes NM, Sharp T. A review of central 5-HT receptors and their function. Neuropharmacology 1999;38:1083-152.
crossref pmid
76. Urban JD, Clarke WP, von Zastrow M, Nichols DE, Kobilka B, Weinstein H, et al. Functional selectivity and classical concepts of quantitative pharmacology. J Pharmacol Exp Ther 2007;320:1-13.
crossref pmid
77. Hoyer D, Clarke DE, Fozard JR, Hartig PR, Martin GR, Mylecharane EJ, et al. International Union of Pharmacology classification of receptors for 5-hydroxytryptamine (Serotonin). Pharmacol Rev 1994;46:157-203.
pmid
78. Nonne C, Lenain N, Hechler B, Mangin P, Cazenave JP, Gachet C, et al. Importance of platelet phospholipase Cgamma2 signaling in arterial thrombosis as a function of lesion severity. Arterioscler Thromb Vasc Biol 2005;25:1293-8.
pmid
79. Chaudhary PK, Kim S, Jee Y, Lee SH, Park KM, Kim S. Role of GRK6 in the regulation of platelet activation through selective G Protein-Coupled Receptor (GPCR) desensitization. Int J Mol Sci 2020;21.
crossref pmid
80. de Chaffoy de Courcelles D, Roevens P, Van Belle H, De Clerck F. The synergistic effect of serotonin and epinephrine on the human platelet at the level of signal transduction. FEBS Lett 1987;219:283-8.
crossref pmid
81. Offermanns S, Toombs CF, Hu YH, Simon MI. Defective platelet activation in G alpha(q)-deficient mice. Nature 1997;389:183-6.
crossref pmid
82. Johnson GJ, Leis LA, Dunlop PC. Specificity of G alpha q and G alpha 11 gene expression in platelets and erythrocytes. Expressions of cellular differentiation and species differences. Biochem J 1996;318:1023-31.
pmid pmc
83. Stephens LR, Eguinoa A, Erdjument-Bromage H, Lui M, Cooke F, Coadwell J, et al. The G beta gamma sensitivity of a PI3K is dependent upon a tightly associated adaptor, p101. Cell 1997;89:105-14.
crossref pmid
84. Vogt S, Grosse R, Schultz G, Offermanns S. Receptor-dependent RhoA activation in G12/G13-deficient cells: genetic evidence for an involvement of Gq/G11. J Biol Chem 2003;278:28743-9.
pmid
85. Brune B, Ullrich V. Calcium mobilization in human platelets by receptor agonists and calcium-ATPase inhibitors. FEBS Lett 1991;284:1-4.
crossref pmid
86. Fraer M, Kilic F. Serotonin. A different player in hypertension-associated thrombosis. Hypertension 2015;65:942-8.
crossref pmid pmc
87. Birk AV, Leno E, Robertson HD, Bolotina VM, Szeto HH. Interaction between ATP and catecholamines in stimulation of platelet aggregation. Am J Physiol Heart Circ Physiol 2003;284:619-25.
crossref pmid
88. Hart MJ, Jiang X, Kozasa T, Roscoe W, Singer WD, Gilman AG, et al. Direct stimulation of the guanine nucleotide exchange activity of p115 RhoGEF by Galpha13. Science 1998;280:2112-4.
crossref pmid
89. Fukuhara S, Chikumi H, Gutkind JS. RGS-containing RhoGEFs: the missing link between transforming G proteins and Rho. Oncogene 2001;20:1661-8.
crossref pmid
90. Kozasa T, Jiang X, Hart MJ, Sternweis PM, Singer WD, Gilman AG, et al. p115 RhoGEF, a GTPase activating protein for Galpha12 and Galpha13. Science 1998;280:2109-11.
crossref pmid
91. Moers A, Nieswandt B, Massberg S, Wettschureck N, Gruner S, Konrad I, et al. G13 is an essential mediator of platelet activation in hemostasis and thrombosis. Nat Med 2003;9:1418-22.
crossref pmid
92. Gong H, Shen B, Flevaris P, Chow C, Lam SC, Voyno-Yasenetskaya TA, et al. G protein subunit Galpha13 binds to integrin alphaIIbbeta3 and mediates integrin “outside-in” signaling. Science 2010;327:340-3.
pmid pmc
93. Clapham DE, Neer EJ. G protein beta gamma subunits. Annu Rev Pharmacol Toxicol 1997;37:167-203.
crossref pmid
94. Hirsch E, Bosco O, Tropel P, Laffargue M, Calvez R, Altruda F, et al. Resistance to thromboembolism in PI3Kgamma-deficient mice. FASEB J 2001;15:2019-21.
crossref pmid
95. Li Z, Zhang G, Le Breton GC, Gao X, Malik AB, Du X. Two waves of platelet secretion induced by thromboxane A2 receptor and a critical role for phosphoinositide 3-kinases. J Biol Chem 2003;278:30725-31.
crossref pmid
96. Kim S, Kunapuli SP. P2Y12 receptor in platelet activation. Platelets 2011;22:56-60.
crossref pmid
97. Dangelmaier C, Kunapuli SP. Evidence for a PI3-kinase independent pathway in the regulation of Rap1b activation downstream of the P2Y12 receptor in platelets. Platelets 2022;33:1301-6.
crossref pmid pmc
98. Chung S, Funakoshi T, Civelli O. Orphan GPCR research. Br J Pharmacol 2008;153:339-46.
crossref pmid pmc
99. Tang XL, Wang Y, Li DL, Luo J, Liu MY. Orphan G protein-coupled receptors (GPCRs): biological functions and potential drug targets. Acta Pharmacol Sin 2012;33:363-71.
crossref pmid pmc
100. Van Doren L, Nguyen N, Garzia C, Fletcher EK, Stevenson R, Jaramillo D, et al. Lipid receptor GPR31 (G-Protein-Coupled Receptor 31) regulates platelet reactivity and thrombosis without affecting hemostasis. Arterioscler Thromb Vasc Biol 2021;41:33-45.

101. Yeung J, Adili R, Stringham EN, Luo R, Vizurraga A, Rosselli-Murai LK, et al. GPR56/ADGRG1 is a platelet collagen-responsive GPCR and hemostatic sensor of shear force. Proc Natl Acad Sci USA 2020;117:28275-86.
crossref pmid pmc
102. Kargl J, Brown AJ, Andersen L, Dorn G, Schicho R, Waldhoer M, et al. A selective antagonist reveals a potential role of G protein-coupled receptor 55 in platelet and endothelial cell function. J Pharmacol Exp Ther 2013;346:54-66.
crossref pmid
103. Sawzdargo M, Nguyen T, Lee DK, Lynch KR, Cheng R, Heng HH, et al. Identification and cloning of three novel human G protein-coupled receptor genes GPR52, PsiGPR53 and GPR55: GPR55 is extensively expressed in human brain. Brain Res Mol Brain Res 1999;64:193-8.
pmid
104. Ryberg E, Larsson N, Sjogren S, Hjorth S, Hermansson NO, Leonova J, et al. The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol 2007;152:1092-101.
crossref pmid pmc
105. Lefkowitz RJ. The superfamily of heptahelical receptors. Nat Cell Biol 2000;2:E133-6.
crossref pmid
106. Kobilka BK. G protein coupled receptor structure and activation. Biochim Biophys Acta 2007;1768:794-807.
crossref pmid pmc
107. Sorkin A, von Zastrow M. Endocytosis and signalling: intertwining molecular networks. Nat Rev Mol Cell Biol 2009;10:609-22.
crossref pmid pmc
108. Pitcher JA, Freedman NJ, Lefkowitz RJ. G protein-coupled receptor kinases. Annu Rev Biochem 1998;67:653-92.
crossref pmid
109. Gurevich EV, Gurevich VV. Arrestins: ubiquitous regulators of cellular signaling pathways. Genome Biol 2006;7:236.
crossref pmid pmc
110. Latorraca NR, Wang JK, Bauer B, Townshend RJL, Hollingsworth SA, Olivieri JE, et al. Molecular mechanism of GPCR-mediated arrestin activation. Nature 2018;557:452-6.
crossref pmid pmc
111. Zeiler M, Moser M, Mann M. Copy number analysis of the murine platelet proteome spanning the complete abundance range. Mol Cell Proteomics 2014;13:3435-45.
crossref pmid pmc
112. Tesmer VM, Kawano T, Shankaranarayanan A, Kozasa T, Tesmer JJ. Snapshot of activated G proteins at the membrane: the Galphaq-GRK2-Gbetagamma complex. Science 2005;310:1686-90.
crossref
113. Hardy AR, Conley PB, Luo J, Benovic JL, Poole AW, Mundell SJ. P2Y1 and P2Y12 receptors for ADP desensitize by distinct kinase-dependent mechanisms. Blood 2005;105:3552-60.
crossref pmid
114. Garcia-Sevilla JA, Alvaro-Bartolome M, Diez-Alarcia R, Ramos-Miguel A, Puigdemont D, Perez V, et al. Reduced platelet G protein-coupled receptor kinase 2 in major depressive disorder: antidepressant treatment-induced upregulation of GRK2 protein discriminates between responder and non-responder patients. Eur Neuropsychopharmacol 2010;20:721-30.
crossref pmid
115. Chen X, Gupta S, Cooper M, DeHelian D, Zhao X, Naik MU, et al. GRK6 regulates the hemostatic response to injury through its rate-limiting effects on GPCR signaling in platelets. Blood Adv 2020;4:76-86.
crossref pmid pmc
116. Shenoy SK, Lefkowitz RJ. Multifaceted roles of beta-arrestins in the regulation of seven-membrane-spanning receptor trafficking and signalling. Biochem J 2003;375:503-15.
pmid pmc
117. Downes K, Zhao X, Gleadall NS, McKinney H, Kempster C, Batista J, et al. G protein-coupled receptor kinase 5 regulates thrombin signaling in platelets via PAR-1. Blood Adv 2022;6:2319-30.
crossref pmid pmc
118. Wu JH, Zhang L, Fanaroff AC, Cai X, Sharma KC, Brian L, et al. G protein-coupled receptor kinase-5 attenuates atherosclerosis by regulating receptor tyrosine kinases and 7-transmembrane receptors. Arterioscler Thromb Vasc Biol 2012;32:308-16.
crossref pmid pmc
119. Gratacap MP, Payrastre B, Nieswandt B, Offermanns S. Differential regulation of Rho and Rac through heterotrimeric G-proteins and cyclic nucleotides. J Biol Chem 2001;276:47906-13.
crossref pmid
120. Gurbel PA, Kuliopulos A, Tantry US. G-protein-coupled receptors signaling pathways in new antiplatelet drug development. Arterioscler Thromb Vasc Biol 2015;35:500-12.
crossref pmid pmc
Editorial Office
Editor-in-Chief: Hun-Gyu Hwang, MD
Department of Internal Medicine, Soonchunhyang University Gumi Hospital,
179 1gongdan-ro, Gumi-si, Gyeongsangbuk-do, 39371, Republic of Korea.
E-mail: hwangpark@schmc.ac.kr

About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Korean Society on Thrombosis and Hemostasis
2714, D-dong, Yongsan Park Xi,
205, Hangang-daero, Yongsan-gu, Seoul 04322, Republic of Korea.
Tel: +82-2-790-2426, Fax: +82-2-790-2429
E-mail: ksth@thrombo.or.kr, http://www.thrombo.or.kr
Copyright© Korean Society on Thrombosis and Hemostasis.              Developed in M2PI