Hamostaseologie 2021; 41(03): 206-216
DOI: 10.1055/a-1476-7873
Review Article

Fine-Tuning of Platelet Responses by Serine/Threonine Protein Kinases and Phosphatases—Just the Beginning

Yavar Shiravand
1   Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
,
Ulrich Walter
2   Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
,
Kerstin Jurk
2   Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
› Author Affiliations
Funding K.J. and U.W. are supported by the German Federal Ministry of Education and Research (BMBF 01EO1003 and 01EO1503).
 

Abstract

Comprehensive proteomic analyses of human and murine platelets established an extraordinary intracellular repertoire of signaling components, which control crucial functions. The spectrum of platelet serine/threonine protein kinases (more than 100) includes the AGC family (protein kinase A, G, C [PKA, PKG, PKC]), the mitogen-activated protein kinases (MAPKs), and others. PKA and PKG have multiple significantly overlapping substrates in human platelets, which possibly affect functions with clear “signaling nodes” of regulation by multiple protein kinases/phosphatases. Signaling nodes are intracellular Ca2+ stores, the contractile system (myosin light chains), and other signaling components such as G-proteins, protein kinases, and protein phosphatases. An example for this fine-tuning is the tyrosine kinase Syk, a crucial component of platelet activation, which is controlled by several serine/threonine and tyrosine protein kinases as well as phosphatases. Other protein kinases including PKA/PKG modulate protein phosphatase 2A, which may be a master regulator of MAPK signaling in human platelets. Protein kinases and in particular MAPKs are targeted by an increasing number of clinically used inhibitors. However, the precise regulation and fine-tuning of these protein kinases and their effects on other signaling components in platelets are only superficially understood—just the beginning. However, promising future approaches are in sight.


#

Introduction

The discovery of adenosine diphosphate (ADP)-induced aggregation of human platelets in 1962[1] enabled subsequent discoveries that platelet-derived thromboxane A2 (TxA2) stimulates[2] and endothelium-derived prostacyclin (PGI2) inhibits[3] [4] platelet activation. Multiple hormones, eicosanoids, and vasoactive factors are now known, which stimulate, inhibit, or modulate major platelet responses such as adhesion, motility, secretion, aggregation, and thrombin generation.[5] [6] [7] Most of these factors act via platelet membrane receptors, which stimulate intracellular signal transduction pathways.[6] [8] [9] [10] Further advances in platelet receptor–mediated signaling developed with the “second messenger concept,” which was originally reported for the effects of glucagon and adrenaline and their second messenger cyclic adenosine monophosphate (cAMP).[11] [12] Two endothelial cell–derived factors, PGI2 and nitric oxide (NO), increase the second messengers cAMP and cyclic guanosine monophosphate (cGMP), respectively, which activate their targets cAMP- and cGMP-dependent protein kinases (PKA, PKG), resulting in platelet inhibition.[3] [4] [10] [13] [14] [15] [16] [17] Additional second messengers including 1,2-diacylglycerol (DAG), inositol 1,4,5-trisphosphate (IP3), phosphoinositides, and Ca2+/calmodulins stimulate distinct serine/threonine protein kinases (ST-PKs), for example, protein kinase C (PKC), calmodulin-dependent protein kinases (CAMKs), myosin light chain kinase (MLCK), and protein kinase B (PKB/Akt), which support platelet activation.[6] [18] Following the discovery of the protooncogene c-src in nondividing human platelets,[19] tyrosine protein kinases (TKs) c-src and other src family kinase (SFK) members (e.g., Lyn, Yes, Fyn, Fgr) were shown to mediate platelet activation in response of glycoprotein (GP) VI, GPIbα, integrins, and CLEC-2.[8] [20] These membrane receptors also affect a sequential kinase cascade, the mitogen-activated protein kinases (MAPKs), in anucleate platelets.[21] A scheme of the major signaling pathways in platelets ([Fig. 1]) illustrates the central role of ST-PKs in human platelets. However, this model, as many other models published, has important limitations. In most cases (as here), they show linear, “receptor-to-function signaling,” which does not reflect the real signaling world within the platelet. In fact, each of the protein kinase shown in this model represents a family of closely related protein kinases, and altogether we have more than 100 platelet ST-PKs ([Supplementary Table S1] [online only]). Each of the kinase systems has multiple protein substrates, often with several phosphorylation sites, which are counterregulated by serine/threonine protein phosphatases (ST-PPs). Furthermore, these protein kinases and phosphatases and their substrates (often other regulatory molecules such as kinases and G-proteins) interact and integrate the signaling response to a functional response, not only at vertical (receptor-to function) level but also at horizontal level of signaling (interaction between various protein kinases). We are only at the beginning to understand this protein kinase network. This and possible future approaches to elucidate this amazing network will be addressed in this review.

Zoom Image
Fig. 1 Multiple intracellular serine/threonine protein kinases mediate the functional responses of platelet membrane receptor stimulation and subsequent second messenger signaling. Numerous physiological and pharmacological platelet activators or inhibitors achieve their effects via specific membrane receptors. G-protein–coupled receptors (GPCRs) activate distinct G-proteins (Gq, Gi/z, Gs), phospholipases C (PLC), and their specific second messenger systems (Ca2+, DAG, PIP3, cAMP) and protein kinase targets. Platelet “pattern-recognition” receptors activate Src family tyrosine kinases (SFK)/immunoreceptor tyrosine-based activation motif (ITAM)/spleen tyrosine kinase (Syk) signaling complexes, which also lead to elevated cytoplasmic levels of second messengers and platelet activation. NO, an endothelium-derived gaseous platelet inhibitor, passes through the platelet membrane, stimulates the soluble guanylyl cyclase (sGC), and increases cytoplasmic cGMP levels. Both the cAMP/PKA and cGMP/PKG systems represent potent inhibitory pathways, which block agonist-induced platelet activation at several steps due to the phosphorylation of multiple substrates. The second messengers Ca2+, DAG, and PIP3 also stimulate specific protein kinases, leading to partially overlapping platelet responses. Similar to PKA/PKG, most of these protein kinases also have multiple substrates, which have been only partially identified. This model shown here and many others published in the field have important limitations as discussed in the text. Platelets actually have hundreds of serine/threonine protein kinases (see [Supplementary Table S1] [online only]) with thousands of substrates and interact with other components such as protein phosphatases to integrate the signaling response to a functional response. This not only occurs at a vertical (receptor-to-function) level, but also at horizontal level of signaling (interaction between various protein kinases). We are only at the beginning to understand this very dynamic and rapidly responding protein kinase network, which is addressed in this review. This model is based on previous models published.[6] [100] [101] [102] [103] [104] 5-HT2A, serotonin receptor; CLEC-2, C-type lectin like-receptor 2; CXCR4/CXCR6/CXCR7, chemokine receptors; DP1, prostanoid receptor D1; EP 2,4, prostanoid receptors EP 2, 4; EP1, EP3, prostanoid receptors EP1 and EP3; GPIb–V–IX, glycoprotein Ib–V–IX complex; GPVI, glycoprotein VI; IP, prostanoid/prostacyclin receptor I; P2Y1, ADP/P2 purinoceptor subtype Y1; P2Y12, ADP/P2 purinoceptor subtype Y12; PAR 1/4, protease-activated receptor 1 and 4; TP, thromboxane receptor; α2A, α2A-adrenergic receptor.

#

ST-PKs in Human and Murine Platelets—AGC (PKA, PKG, PKC)

The elucidation of the complete sequence of the human genome combined with bioinformatic approaches defined the entire PK and PP complement of the human genome, kinome, and phosphatome, respectively.[22] [23] This fundamental step in signaling research established, based on their catalytic domain, the presence of 518 PK genes (+ 106 pseudogenes) and 189 PP genes (+ 79 pseudogenes) in humans. However, the genomic presence of PKs and PPs does not indicate that these proteins are expressed at the protein level. Advances in proteomics enabled comprehensive and quantitative analyses of the human and murine platelet proteome, including their major signaling components.[24] [25] Both human and murine platelets express a large spectrum of PKs from essentially all classes.[24] [25] [26] Here, the quantitative expression of human and murine platelet ST PKs is compared for the first time ([Supplementary Table S1] [online only]) together with a short update on the AGC protein kinase families (PKA/PKG/PKC). Then, specific examples of novel interactions between ST-PKs, TKs, and ST-PPs are discussed followed by a short update on platelet MAPKs. The quality and similarity of the original human and murine platelet proteomic data obtained by two different groups suggest that the protein kinase spectrum observed represent true positive results. On the other hand, it is known that a global proteomic approach may miss certain proteins, especially membrane proteins and proteins with very low expression. Therefore, platelets may have some additional protein kinases, which are not listed in our [Supplementary Table S1] (online only).

Human platelets express with more than 110 ST PKs approximately 25% of the human kinome (518 known PKs), in particular the PK families PKA, PKG, PKC, and CAMK.[22] [27] PKA is the most extensively characterized kinase of this group. Under basal conditions, PKA is a hetero-tetrameric, inactive holoenzyme and consists of two regulatory and two catalytic subunits. With cAMP elevation, this cyclic nucleotide binds to the two regulatory subunits of the holoenzyme, causes a conformational change, and releases active catalytic subunits. PKA was the first example of a PK activated by second-messenger–induced loss of autoinhibition. Other mechanisms include the activation by phosphorylation/autophosphorylation ([Supplementary Table S1] [online only]), which often also release autoinhibition. The human genome codes for seven different PKA subunits: three different catalytic subunits (PRKACA, PRKACB, PRKACG), two different regulatory subunits type I (PRKAR1A, PRKAR1B), and two different regulatory subunit type II (PRKAR2A, PRKAR2B). The main isoforms of regulatory and catalytic subunits expressed in human platelets are R1A, R1B, R2B, CA, and CB, which may form several distinct PKA holoenzymes.[16] [24] However, very little is known regarding the contribution of the individual PKA subunits. PKA regulatory subunits influence the spatiotemporal PKA activation, due to RI/RII-specific A-kinase anchoring proteins (AKAPs).[28] [29] [30] [31]

PKG is evolutionary, structurally and functionally closely related to PKA, but comprises a single polypeptide chain, containing both catalytic and two regulatory (cGMP-binding) domains, and there are two distinct forms, a soluble (type I) and a membrane-bound form (type II).[32] The human gene PRKG1 (chromosome 10) encodes the soluble Iα/Iß isoforms produced by alternative transcript splicing, another gene on human chromosome 4, PRKG2, encodes the membrane-bound PRKG isoform II. Human and murine platelets do not express PKG type II, but PKG Iß at high (human) or low (murine) levels as shown in [Supplementary Table S1] (online only).[16] [24]

Although there are other proteins, which specifically bind cAMP and/or cGMP, multiple lines of genetic, biochemical, physiological, and pharmacological evidence revealed that the inhibitory effects of cAMP- and cGMP-elevating agents on platelet activation and function are mediated by PKA and PKG, respectively, as reviewed elsewhere.[10] [16] [17] The platelet inhibitory effects of cGMP-elevating agents or membrane permeable cGMP analogs are strongly impaired in human or murine platelets lacking PKG I, whereas the cAMP/PKA system was not affected.[14] [33] Previously, a small numbers of PKA and/or PKG substrates were identified in platelets such as G-proteins/G-protein-associated proteins, cytoskeletal proteins, proteins associated with intracellular Ca2+-stores/-release, cell membranes, and cAMP/cGMP phosphodiesterases.[16] [34] Selective stimulation of the human platelet cAMP/PKA system (by the PGI2 analog iloprost) or of platelet PKG (by the soluble guanylyl cyclase stimulator riociguat) increased the phosphorylation of 299 (iloprost) and more than 150 (riociguat) proteins, and decreased the phosphorylation of more than 60 proteins (with riociguat).[10] [35] [36] [37] Surprisingly, there was a large but not complete overlap between the PKA- and PKG-regulated phosphoproteins, in agreement with the earlier conventional studies.[10] [16] This PKA/PKG overlap raises the hypothesis that each pathway may be, at least partially, a back-up for each other. Notably, there are cellular clusters (signaling nodes) with several PKA/PKG-regulated phosphoproteins, which often contain multiple phosphosites.[10] [17] [38] Remarkably, both PKA and PKG affect the phosphorylation of other PKs and PPs, as demonstrated with two specific examples, the TKs Src and Syk, and the protein phosphatase 2A (PP2A).[35] [36]

Following cAMP/cGMP pathways, other second messengers such as diacylglycerol (DAG), inositol-3-phosphate (IP3), Ca2+, phospholipids, and their target protein kinases PKC and MLCK were discovered. Ca2+/calmodulin-dependent MLCK[39] [40] [41] and PKC[42] [43] [44] are closely associated with platelet activation, in particular shape change and secretion. MLCK phosphorylates myosin light chain (MLC) at S18/T19 and increases the contractile force of the myosin–F-actin complex, which is reversed by myosin light chain phosphatase (MLCP)-mediated MLC dephosphorylation. This MLCK–MLC–MLCP complex is tightly regulated by multiple PKs and PPs. MLCK is activated by Ca2+/calmodulin binding, by TK- and PKC-mediated phosphorylation, but is inhibited by PKA and PKG, whereas MLCP is inhibited by Rho-associated protein kinase (ROCK)-mediated phosphorylation.[45] [46] In platelets, many components of the MLCK–MLC–MLCP complex are major “signaling nodes” of motility regulation and phosphorylated at multiple sites by both PKA and PKG, resulting in decreased MLC phosphorylation and reduced contractile force.[10] [47] [48]

The classical PKC is one protein chain with a C-terminal kinase domain and N-terminal regulatory moieties, which bind the activating second messengers DAG and Ca2+.[18] [49] The tumor promotor phorbol ester is a potent activator of PKCs, binds to the DAG regulatory site, and is an excellent pharmacological tool to activate PKC. The entire PKC family consists of the classical/conventional forms (α, βI, βII, γ), novel forms (δ, θ, ε, ŋ), and atypical forms (ζ, ɩ/λ) with conserved catalytic domains, but different regulatory moieties.[44] Human and mouse platelets express PKC α, β, δ, and θ at detectable levels, while the expression of PKCŋ is very low and PKCε is restricted to mouse platelets.[18] [50] In vivo studies demonstrated that PKCα-deficient mice show a reduced phosphorylation of synaptosomal-associated protein 23 (SNAP-23, part of the soluble N-ethylmaleimide-sensitive factor attachment receptor [SNARE] complex) at S95, associated with decreased α-granule secretion and impaired dense granule biogenesis. Platelets from PKC δ, θ, and ε knockout mice exhibited impaired phosphorylation of vasodilator-stimulated phosphoprotein, pleckstrin, and syntaxin-4, respectively.[50] PKC isoforms (α and β) appear to have nonredundant roles in regulating α-granule secretion, integrin activation, and Ca2+ signaling, whereas PKCδ and θ negatively regulate thrombus formation on collagen.[18] [50] [51] PKCδ is suggested to be involved in collagen-induced platelet activation via the synthesis of TxA2, mediated at least in part by mitogen-activated protein kinase kinase (MAP2K/MEK)/extracellular signal-regulated kinase (ERK)/p38 MAPK pathways.[52] The observation that members of the PKC family have opposing functional roles in platelets suggests that these differences are due to distinct substrates, which need to be elucidated.[18] [50]


#

Tyrosine (Y) PKs as Targets of ST-PKs and ST-PPs: Fine-Tuning of Syk

Various adhesion molecules activate platelets by binding to distinct transmembrane receptors (GPVI, GPIbα, CLEC-2, and αIIbβ3 integrin). Activation of intracellular, membrane-associated Src family tyrosine kinases (SFKs) increases Y-phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAM) and subsequent membrane recruitment and stimulation of the spleen Y kinase (Syk).[8] [53] Recruitment of Syk to ITAM at the membrane via its two Src-homology 2 (SH2) domains and Y-phosphorylation of the sites Y352 (by SFKs) and Y525/526 (by autophosphorylation) abolishes Syk autoinhibition ([Fig. 2]).[54] [55] [56] This sophisticated SFK-Syk cascade activates phospholipase (PLC) γ2, via Y-phosphorylation, leading to elevated DAG/IP3/Ca2+ and PKC activation.[53] Syk also stimulates ADP secretion and TxA2 synthesis, which enhance initial platelet activation. Syk is the only ppITAM-activated TK in platelets and therefore represents an essential component of adhesion-dependent platelet activation. Many TKs are known to be phosphorylated by ST-PKs, but the functional role is often not known.

Zoom Image
Fig. 2 Model showing activation, inhibition, and fine-tuning of adhesion-dependent Syk regulation in human platelets. Activated GPVI and GPIbα increase SFK-mediated dual phosphorylation of FcRγ ITAM motifs (ppITAM), which recruit inactive Syk (via its SH2 domains) from the cytosol to the cell membrane. This induces a conformational change leading to Syk activation and concomitant Y-phosphorylation of the interdomain B (Y352) and kinase domain (Y525/6), mainly by SFKs and autophosphorylation, respectively. Fully activated Syk and Syk interacting proteins such as LAT, Btk, PLCγ2, E3 ubiquitin-protein ligase CBL form a signalosome complex to trigger various cellular responses. Often, Syk tyrosine phosphorylates its interacting proteins in such complexes. GPVI ligands such as collagen, VWF, and convulxin also induce a Syk- and PKC-mediated S-phosphorylation (S297) within the interdomain B of Syk, which is negatively associated with Y-phosphorylation (Y352, Y525, Y526). Inhibition of the serine/threonine protein phosphatase PP2A by OA also results in Syk S297 phosphorylation due to inhibited dephosphorylation, introducing PP2A as novel factor for Syk regulation. PKA inhibits PKC-mediated Syk S297 phosphorylation at the level of generation and/or action of the secondary mediators ADP and TxA2. The tyrosine phosphatase TULA-2 downregulates Syk tyrosine phosphorylation (mainly pY352) and activity. Syk and its interdomain B are “signaling nodes” for the regulation by multiple serine/threonine and tyrosine protein kinases and phosphatases. ADP, adenosine diphosphate; Btk, Bruton tyrosine kinase; cAMP, cyclic adenosine monophosphate; Cbl, casitas B-lineage lymphoma; cGMP, cyclic guanosine monophosphate; DAG, diacylglycerol; FcRγ, Fc receptor γ-chain; GPCRs, G-protein–coupled receptors; GPVI, glycoprotein VI; IP3, inositol trisphosphate; LAT, linker for activation of T-cells; OA, okadaic acid; PI3K, phosphatidylinositol 3-kinase; PKA, protein kinase A; PKC, protein kinase C; PLCγ2, phospholipase Cγ2; PP2A, protein phosphatase 2A; SFK, src kinase family; SLP-76, Src homology (SH) 2 domain-containing leukocyte phosphoprotein of 76 kDa; Syk, spleen tyrosine kinase; TULA-2, T-cell ubiquitin ligand-2; TxA2, thromboxane A2; VWF, von Willebrand factor.

Interestingly, in our global phosphoproteomic studies with platelets, the cAMP/PKA pathway affected the phosphorylation of many protein kinases, including the TKs Src and Syk.[10] [35] [36] The observed cAMP/PK-stimulated Src Y530 phosphorylation is certainly not a direct PKA phosphosite and likely to be mediated by PKA phosphorylation of C-terminal Src kinase (CSK) at S364. This stimulates CSK to phosphorylate inhibitory tyrosine sites (e.g., Y530) at the C-terminus of Src and other SFKs.[57] [58] [59] This is one example that PKA affects a TK cascade (CSK, SFK, Syk) and their downstream targets. A recent study, using a different analytical approach (fluorescence two-dimensional gel electrophoresis) also reported that the cAMP/PKA system increases the phosphorylation status of several signaling components including protein phosphatase 1A (PPA1), Src kinase-associated phosphoprotein 2 (SKPA2), cGMP-dependent protein kinase 1 (PRKG1), integrin-linked protein kinase (ILK), and the dual specificity protein phosphatase 3 (DUSP3).[60] However, this method does not identify the identity of the phosphorylation sites, and the results are therefore not directly comparable to other mass spectrometry-based phosphoproteomic studies.

The PKA/PKG pathways downregulated basal and strongly inhibited ADP-stimulated Syk S297 phosphorylation in intact human platelets.[36] [61] The S297 phosphosite is located within the Syk interdomain B, which maintains this TK in an autoinhibited state under basal conditions and participates in conformation-dependent Syk activation upon ITAM-binding and Y-phosphorylation as shown in [Fig. 2].[54] [55] [56]

GPVI-/GPIbα-mediated platelet activation increased the well-established Syk Y-phosphorylation/activation and the stoichiometric, transient PKC-mediated Syk S297 phosphorylation as well. PKC inhibition abolished this Syk S297 phosphorylation, but enhanced GPVI-/GPIbα-increased Syk Y-phosphorylation/activity, indicating a possible feedback inhibition.[62] Furthermore, inhibition of the ST PP2A alone increased Syk S297 phosphorylation without affecting Syk Y-phosphorylation and activity, whereas PP2A inhibition preceding GPVI-mediated platelet activation downregulated Syk Y-phosphorylation and activity.[63] [Fig. 2] summarizes that Syk, an established central component of adhesion-dependent platelet activation, is controlled and fine-tuned by multiple Y- and ST-PKs as well as by corresponding protein phosphatases. Upon activation, Syk interacts with Y-phosphorylates and activates further signaling components such as PLCγ2 ([Fig. 2]). Furthermore, there are many more Syk substrates known in platelets and other Syk–containing cells such as B-cells, lymphoma, and breast cancer cells.[64] [65] These developments lead to the recognition that Syk can function as an oncogene or tumor suppressor, depending on the cellular context.[66] Presently, there are major efforts ongoing to elucidate the Syk signaling network by functional and phosphoproteomic analyses combined with bioinformatic network modeling.[65] It is very likely that these approaches can also be successfully applied to human platelets in the future.


#

ST-PPs as Targets of ST-PKs in Human Platelets

PPs reverse PK-mediated phosphorylations and are encoded by 189 PP genes in humans, based on their catalytic subunits.[23] However, the number of PP holoenzymes is much larger than the number of catalytic subunits due to additional regulatory subunits. PPs are classified into 10 different protein folds and 21 families, including Y-PPs (largest group) and ST-PPs (second largest group) as major families, and Y- and ST-PPs are widely expressed in human and murine platelets.[24] [25] [26] Four different dual-specificity (Y + ST) PPs (DUSPs) and 15 protein Y-phosphatases (PTPs) were detected, including some receptor Y-PP.[26] [67] Platelet Y-PPs represent an important signaling system with a therapeutic potential as well reviewed by others.[67] [68]

Human ST-PPs contain two major families, metallo-dependent PPs (PPM fold) and phosphoprotein phosphatases (PPP/PPPL fold, with subfamilies PPP 1–6), which are well expressed in human platelets.[69] The (P)PP1 subfamily regulates contraction in smooth muscle and platelets, as it reverses the action of MLC kinase (MLCK). This family is characterized by three distinct catalytic (α, β, γ) and sixteen different regulatory subunits.[46] Our platelet proteomic studies identified three catalytic subunits and seven different regulatory subunits of PP1, including the gene product PPP1R12A, the myosin phosphatase targeting subunit 1 (MYPT1). MLCP, a special PP1 holoenzyme, is a trimeric enzyme containing a catalytic subunit, a regulatory subunit of uncertain function, and MYPT1, which targets the MLCP holoenzyme to the MLC/myosin complex.[70] The regulatory switch of myosin contractility is the state of MLC phosphorylation, tightly controlled by the balance of MLCK and MLCP, which are themselves regulated by multiple mechanisms and multisite phosphorylation (signaling nodes).

The second major ST-PP, PP2A, has also been known for decades, but its structural and functional diversity was only more recently appreciated,[46] [71] also in human platelets.[26] [69] PP2A is expressed as heterotrimeric holoenzyme composed of a catalytic subunit (C), a scaffold subunit (A) and a targeting/regulatory subunit (B). In humans, A- and C-subunits of PP2A each have two variants (α, β), whereas B-subunits are encoded by 15 different genes, resulting in at least 23 isoforms due to alternative splicing. Based on this, human cells may contain up to 92 different trimeric PP2A holoenzymes. Previously, we presented an overview of PP2A heterogeneity in human platelets.[26]

Elegant studies with Xenopus oocytes, Drosophila, and mammalian cells established that cell mitosis is tightly controlled by multiple phosphorylation steps and a balance between PK and PP activities, in particular by the master mitotic cyclin-dependent kinase 1 (Cdk1) and its antagonizing PP, PP2A-B55. This balance is markedly altered, when PP2A-B55 is strongly inhibited by two regulatory phosphoproteins, α-endosulfine (ENSA) and cAMP-regulated phosphoprotein 19 (ARPP19). For this function, these proteins need to be phosphorylated at a centrally located, highly conserved FDSpGDY motif by Great wall kinase (Gwl; microtubule-associated serine/threonine kinase like [MASTL] in humans).[72] [73] [74] This Gwl–Arpp19–ENSA–PP2A-B55 pathway plays an essential role in the control of the cell cycle from yeast to human, but is thought to have also other functions.[74] Recently, we reported the presence and intracellular concentration of a larger spectrum of ST-PPs in human platelets and addressed in particular the diversity of the PP2A family.[69] Both human and murine platelets contain multiple PP2A subunits (two catalytic, one scaffolding, and seven regulatory) and two endogenous PP2A inhibitors, ENSA and ARPP19.[69] Furthermore, the entire MASTL(like)–ENSA/ARPP19–PP2A pathway was found to be present in human platelets and affected by both PKA and PKG.[69] Exciting questions concern the initiation of this novel pathway in human platelets and its phosphoprotein targets, which are expected to be different from the Cdk cell cycle system. Inhibition of PP2A in human platelets was found to increase the phosphorylation of several MAPKs, indicating their activation.[69] This is in line with the proposed PP2A function as tumor suppressor, as tumor cells often have a reduced and/or impaired PP2A activity resulting in increased MAPKs.[75] Considerable efforts were therefore undertaken in this field for some time to “repair” or elevate selective PP2A forms in diseases. These efforts were very recently rewarded when two major groups demonstrated that some novel phenothiazine derivatives (also called “small molecule activator of PP2A,” SMAP) reactivate specific PP2A isozymes with potential benefit in cancer and other diseases.[76] [77] [78]

It is still a major challenge to elucidate the PP2A isoform-specific signaling and their precise roles on platelet functions, including the observed effects on MAPK signaling. However, this will be aided now by isoform-specific endogenous inhibitors such as ENSA/ARPP19[69] and by novel, selective pharmacological PP2A activators.[78]


#

MAPKs and Their Interactions in Platelets

Recent proteome studies revealed that human and murine platelets abundantly express multiple members of the MAPK family, which belong to the CMGC (CDK, MAPK, GSK3 [glycogen synthase kinase 3], CDK-like kinase) class of serine/threonine kinases[24] [25] ([Supplementary Table S1] [online only]). The canonical MAPK cascades are initiated by a variety of platelet receptor signaling ([Fig. 3]). PK-induced phosphorylation or small GTPase interaction lead to the activation of MAP kinase kinase kinase kinases (MAP4K, MEKKK) and MAP kinase kinase kinases (MAP3K, MEKK), which in turn activate MAP kinase kinases (MAP2K, MEK). Active MAP2Ks then activate MAPKs through dual phosphorylation of threonine/tyrosine residues within the activation loop of the kinase domain. The ERK1/2 (MAPK3/MAPK1) and ERK5 (MAPK7), the c-Jun N-terminal kinase JNK1/2 (MAPK8/MAPK9), and the p38 isoforms α and β (MAPK14) represent prominent MAPKs in human and murine platelets ([Fig. 3]).

Zoom Image
Fig. 3 Signaling network of MAPK in human and murine platelets. Black and red lines demonstrate common activatory and inhibitory signaling pathways in human and murine platelets, respectively. Mouse- and human-specific signaling pathways are shown in blue and gray, respectively. (Modified from various studies.[21] [79] [82] [105] [106] [107]) AA, arachidonic acid; ADP, adenosine diphosphate; APP, amyloid precursor protein; ASK1 (MAP3K5), apoptosis signal-regulating kinase 1; JNK1/2, c-Jun N-terminal kinase 1/2; p38 (MAPK14), mitogen-activated protein kinase 14; cPLA2, cytosolic phospholipases A2; ERK 1/2 (MAPK3/MAPK1), mitogen-activated protein kinase 3 and 1, extracellular signal-regulated kinase 1 and 2; FcRγ, Fc receptor γ-chain; JNK1/2 (MAPK 8 and 9), mitogen-activated protein kinase 8 and 9; MAP3K3, mitogen-activated protein kinase kinase kinase 3; MEK (MAP2K) 1/2, 5, MAP kinase kinases 1/2 and 5; MINK1 (MAP4K6), Misshapen-like kinase 1; MK2, MAPK-activated protein kinase 2; MKK (MAP2K), MAP kinase kinases; NLRP3, NLR family pyrin domain containing 3; NOX, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase; oxLDL, oxidized low-density lipoprotein; PARs, protease-activated receptors; PKC, protein kinase C; PP2A, protein phosphatase 2A; RAF, rapidly accelerated fibrosarcoma; ROS, reactive oxygen species; Syk, spleen tyrosine kinase; TP, thromboxane receptor; TxA2 receptor; TxA2, thromboxane A2; VWF, von Willebrand factor.

Despite advanced platelet MAPK research in the last years, the identification of proximal MAP3/4Ks, their underlying activation mechanisms, and signaling networks are still not well understood, although several members are present ([Supplementary Table S1] [online only]). Murine platelets deficient in the MAP4K misshapen/Nck-interacting kinase–related kinase 1 (MINK1) are characterized by diminished MEK1/2, MKK3/6, ERK, and p38 phosphorylation, associated with decreased δ-granule release of ADP, especially in response to low concentrations of thrombin and collagen. This secretion defect might explain the prolonged bleeding time and impaired thrombus formation under arterial shear conditions in vitro and in vivo[79] ([Fig. 3]). The impact of MINK1 on human platelet function has yet to be proven. Although the MAP3K3 (MEKK3) was not detected in human or murine platelets by proteomics studies,[24] [25] there is evidence from MAP3K3-deficient mice that it regulates JNK1/2 and ERK1/2 activation.[80] The comprehensive identification and functional characterization of JNK1/2 and ERK1/2 substrates also bear challenges. Agonist-induced phosphorylation of cytosolic phospholipase A2 (cPLA2) at S505, representing cPLA2 activity and leading to TxA2 synthesis, was not affected in MAP3K3-deficient platelets,[80] whereas the MEK1/2 inhibitors PD184354 and U0126, used for the analysis of ERK1/2 regulation, clearly diminished cPLA2 S505 phosphorylation and TxA2 synthesis in stimulated human platelets.[81] [82] As these inhibitors are also known to inhibit cyclooxygenase-1 and TxA2-synthase, these off-target effects might be responsible for the conclusion that ERK1/2 is a regulator of TxA2 generation in platelets ([Table 1]). Recently, another MAP3K, apoptosis signal-regulating kinase 1 (ASK1; MAP3K5), was identified to be crucially involved in hemostasis and arterial thrombosis in murine platelets.[83] In ASK1-deficient platelets, MKK3/6, MKK4, and p38 phosphorylation was decreased, cPLA2 was not phosphorylated at S505, and decreased TxA2 formation was observed in line with impaired platelet functions induced by submaximal agonist concentrations. ASK1 deficiency was recapitulated by using the pharmacological ASK1 inhibitor selonsertib in murine wild-type platelets. Conversely, selonsertib in human platelets only blocked early phosphorylation of p38 and its substrate MAPKAP-kinase 2 (MK2, MAPKAPK2), but not sustained phosphorylation, and neither cPLA2 phosphorylation, TxA2 formation, nor agonist-induced platelet activation was reduced.[82] These studies clearly demonstrate that MAPK signaling may differ between murine and human platelets, and effects of pharmacological MAPK inhibitors have to be validated in both species with respect to their specific signaling and potential off-target effects. A recent comprehensive overview summarizes our current knowledge of MAPK signaling and function in murine and human platelets[21] ([Fig. 3], [Table 1]).

Table 1

Off-target and functional in vitro effects of pharmacological MAP kinase inhibitors on human platelets

Inhibitor (target)

Inhibitor (off-target effects)

Functional effects in vitro[a]

ERK1/2

U0126 (MEK1/2)

PD98059 (MEK1/2)

PD184161 (MEK1/2)

PD184352 (MEK1/2)

Cobimetinib (MEK1/2)

U0126 (MEK5)

PD98059 (COX-1/2; TBXAS1; MEK5)

PD184161 (NR)

PD184352 (MEK5)

Cobimetinib (NR)

Adhesion (venous, arterial shear)

TxA2 synthesis

Agonist-specific granule secretion

αIIbβ3 activation/aggregation

Clot retraction

JNK1/2

SP600125 (JNK1/2/3)

6o (JNK1/2/3)

SP600125 (e.g., SGK; S6K1)

6o (NR)

Adhesion (arterial shear)

α-Granule secretion (submaximal agonist concentrations)

αIIbβ3 activation/aggregation

p38

SB203580 (p38α/β)

SB202190 (p38α/β)

Skepinone-L (p38α/β)

VX-702 (p38α/β)

SB203580 (COX-1/2; TBXAS1)

SB202190 (NR)

Skepinone-L (NR)

VX-702 (NR)

Adhesion (static, venous/arterial shear conditions)

TxA2 synthesis

Granule secretion

αIIbβ3 activation/aggregation

Clot retraction

ERK5

XMD8–92 (ERK5)

BIX02189 (MEK5; ERK5)

XMD8–92 (e.g., TNK1; Pik4)

BIX02189 (NR)

Substrate-specific adhesion (arterial shear)

Agonist-specific αIIbβ3 Activation/aggregation

Agonist-specific PS exposure

Abbreviations: COX 1/2, cyclooxygenase 1/2; ERK1/2, extracellular signal-regulated kinase 1/2; JNK1/2, c-Jun N-terminal kinase 1/2; MEK 1/2, mitogen-activated protein kinase kinase; NR, not reported; Pik4, polo-like kinase 4; PS, phosphatidylserine; S6K1, ribosomal protein S6 kinase β-1; SGK, serum and glucocorticoid inducible protein kinases; TBXAS1, thromboxane A synthase 1; TNK1, nonreceptor tyrosine-protein kinase 1; TxA2, thromboxane A2.


Source: Modified from Patel and Naik.[21]


a Off-target effects cannot be excluded.


Interestingly, the crosstalk between MAPKs and S/T-PPs is insufficiently known, too. Thrombin-induced ERK1/2 phosphorylation is increased by ASK1 or p38 inhibitors in human platelets,[82] which was also observed for murine platelets.[83] [84] Potentially, a crosstalk between p38 and S/T-PPs such as PP2A might be responsible for the negative regulation of ERK1/2 by p38.[85] In accordance with these data, we showed that specific inhibition of PP2A by okadaic acid results in increased phosphorylation of ERK1/2 but also of p38 in human platelets, suggesting that PP2A represents an important regulator of distal platelet MAPKs[69] ([Fig. 3]).

Clinically established cancer therapeutics targeting Y-PKs such as Syk or Btk have been intensively analyzed regarding their antiplatelet properties due to their bleeding risk.[86] [87] [88] Interestingly, current clinical trials for the approvement of MAPK inhibitors as anticancer drugs revealed that MEK1/2 (ERK1/2) inhibitors are associated with higher bleeding risk than p38 inhibitors.[21] Much less is known about platelet effects of clinically used anticancer drugs targeting the S/T-PP PP2A.[89] [90] In this regard, it is important to note that novel therapeutics targeting more specifically MAPKs and PP2A may have unwarranted, off-target but also beneficial effects on platelets, megakaryopoiesis, and platelet production. There is evidence that the MEK1/2-ERK1/2 pathway is important for the physiological regulation of megakaryocyte maturation and proplatelet production induced by thrombopoietin via c-MPL (myeloproliferative leukemia virus oncogene)-receptor/JAK2 (janus kinase-2) signaling.[91] [92] On the other hand, inherited ankyrin repeat domain-containing protein 26 (ANKRD26)-related thrombocytopenia is associated with defective proplatelet formation, but with persistent megakaryocyte MEK1/2-ERK1/2 activation and ANKRD26 expression. Interestingly, the MAPK inhibitor PD98059 could correct impaired proplatelet formation of the patients' megakaryocytes.[93] Accordingly, persistent p38-MAPK activation induced by Flt3 (fms-related receptor tyrosine kinase 3)-mediated pathways independent of JAK2 seems to be involved in the control of dysmegakaryopoiesis in primary myelofibrosis.[94] A recent study analyzing MASTL-mutant mice suggested that a distinct pathogenic gain-of-function variant in MASTL related to autosomal dominant thrombocytopenia, E167D, mediates decreased activity of PP2A-B55, leading to hyperphosphorylation of multiple actin cytoskeleton components, hyperstabilized pseudopods, and actin polymerization defects.[95]


#

Conclusion and perspective

Our understanding of Y-PKs and MAPK regulation by ST-PKs and ST-PPs in platelets is just beginning. Their crosstalk is not characterized by linear pathways from “receptor-to-function signaling,” but has to be considered as complex network. Multiple PKs, PPs of different families, and their regulatory substrates interact and integrate signaling processes to a functional outcome. Only multi-pathway interactions between the Y-PK Syk and ST-PKs as well as ST-PP promote a fine-tuned regulation of platelet Syk activation/activity, emphasizing the crucial role of Syk in “pattern-recognition receptor”-mediated hemostatic, inflammatory, and immune functions of platelets. In the future, there is a need for advanced phosphoarrays and/or mass spectrometry-based quantitative phosphoproteomics combined with sophisticated bioinformatics and function tools for high-throughput analyses to establish a deep profile of platelet signaling–function relations. Is this far away and/or unrealistic? Recently, rapid and site-specific deep phosphoproteome profiling by data-independent acquisition without the need for spectral libraries was reported and successfully applied to characterize the epidermal growth factor signaling pathway of two human cell lines.[96] Clinical application is not too far away since the profile of hundreds of phosphoproteomes could be obtained in a few days from a starting protein material as low as 200 µg or less. Interestingly, the signaling pathways in this study were validated by a panel of 30, highly selective protein kinase inhibitors. Since the first successful use of a protein kinase inhibitor (imatinib/ Gleevec) to treat leukemia patients in 2001, 52 FDA-approved small-molecule protein kinase inhibitors are listed as of 2020 for a diverse spectrum of therapeutic indications, among them also the Syk inhibitor fostamatinib (Tavalisse) to treat chronic immune thrombocytopenia.[97] PK inhibitors and more recently drugs targeting PPs have come a long way, are now target-designed, and powerful reagents to control and repair diseased signal transduction pathways. However, their targeted pathways need to be understood and assessed with valid assays. Such emerging strategies[65] [98] [99] are promising for the individualized screening of platelet-related diseases and drugs. Human platelets can serve here as prominent cellular model, but also as therapeutic and diagnostic target.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Supplementary Material

  • References

  • 1 Born GV. Aggregation of blood platelets by adenosine diphosphate and its reversal. Nature 1962; 194: 927-929
  • 2 Svensson J, Hamberg M, Samuelsson B. On the formation and effects of thromboxane A2 in human platelets. Acta Physiol Scand 1976; 98 (03) 285-294
  • 3 Moncada S, Higgs EA, Vane JR. Human arterial and venous tissues generate prostacyclin (prostaglandin x), a potent inhibitor of platelet aggregation. Lancet 1977; 1 (8001): 18-20
  • 4 Tateson JE, Moncada S, Vane JR. Effects of prostacyclin (PGX) on cyclic AMP concentrations in human platelets. Prostaglandins 1977; 13 (03) 389-397
  • 5 Jurk K, Kehrel BE. Platelets: physiology and biochemistry. Semin Thromb Hemost 2005; 31 (04) 381-392
  • 6 Versteeg HH, Heemskerk JWM, Levi M, Reitsma PH. New fundamentals in hemostasis. Physiol Rev 2013; 93 (01) 327-358
  • 7 Jurk K. Analysis of platelet function and dysfunction. Hamostaseologie 2015; 35 (01) 60-72
  • 8 Senis YA, Mazharian A, Mori J. Src family kinases: at the forefront of platelet activation. Blood 2014; 124 (13) 2013-2024
  • 9 Brass LF, Ma P, Tomaiuolo M. et al. A systems approach to the platelet signaling network and the hemostatic response to injury. In: Platelets in Thrombotic and Non-Thrombotic Disorders. Springer; 2017: 367-378
  • 10 Makhoul S, Walter E, Pagel O. et al. Effects of the NO/soluble guanylate cyclase/cGMP system on the functions of human platelets. Nitric Oxide 2018; 76: 71-80
  • 11 Sutherland EW, Robison GA. The role of cyclic-3′,5′-AMP in responses to catecholamines and other hormones. Pharmacol Rev 1966; 18 (01) 145-161
  • 12 Sutherland EW. On the biological role of cyclic AMP. JAMA 1970; 214 (07) 1281-1288
  • 13 Mellion BT, Ignarro LJ, Ohlstein EH, Pontecorvo EG, Hyman AL, Kadowitz PJ. Evidence for the inhibitory role of guanosine 3′, 5′-monophosphate in ADP-induced human platelet aggregation in the presence of nitric oxide and related vasodilators. Blood 1981; 57 (05) 946-955
  • 14 Eigenthaler M, Ullrich H, Geiger J. et al. Defective nitrovasodilator-stimulated protein phosphorylation and calcium regulation in cGMP-dependent protein kinase-deficient human platelets of chronic myelocytic leukemia. J Biol Chem 1993; 268 (18) 13526-13531
  • 15 Waldmann R, Bauer S, Göbel C, Hofmann F, Jakobs KH, Walter U. Demonstration of cGMP-dependent protein kinase and cGMP-dependent phosphorylation in cell-free extracts of platelets. Eur J Biochem 1986; 158 (01) 203-210
  • 16 Smolenski A. Novel roles of cAMP/cGMP-dependent signaling in platelets. J Thromb Haemost 2012; 10 (02) 167-176
  • 17 Nagy Z, Smolenski A. Cyclic nucleotide-dependent inhibitory signaling interweaves with activating pathways to determine platelet responses. Res Pract Thromb Haemost 2018; 2 (03) 558-571
  • 18 Kunapuli SP, Bhavanasi D, Kostyak JC. et al. Platelet signaling: protein phosphorylation. In: Platelets in Thrombotic and Non-Thrombotic Disorders: Pathophysiology, Pharmacology and Therapeutics: An Update. Springer International Publishing; 2017: 297-308
  • 19 Golden A, Nemeth SP, Brugge JS. Blood platelets express high levels of the pp60c-src-specific tyrosine kinase activity. Proc Natl Acad Sci U S A 1986; 83 (04) 852-856
  • 20 Séverin S, Nash CA, Mori J. et al. Distinct and overlapping functional roles of Src family kinases in mouse platelets. J Thromb Haemost 2012; 10 (08) 1631-1645
  • 21 Patel P, Naik UP. Platelet MAPKs-a 20+ year history: What do we really know?. J Thromb Haemost 2020; 18 (09) 2087-2102
  • 22 Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science 2002; 298 (5600): 1912-1934
  • 23 Chen MJ, Dixon JE, Manning G. Genomics and evolution of protein phosphatases. Sci Signal 2017; 10 (474) 10
  • 24 Burkhart JM, Vaudel M, Gambaryan S. et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 2012; 120 (15) e73-e82
  • 25 Zeiler M, Moser M, Mann M. Copy number analysis of the murine platelet proteome spanning the complete abundance range. Mol Cell Proteomics 2014; 13 (12) 3435-3445
  • 26 Jurk K, Walter U. New Insights into platelet signalling pathways by functional and proteomic approaches. Hamostaseologie 2019; 39 (02) 140-151
  • 27 Miranda-Saavedra D, Barton GJ. Classification and functional annotation of eukaryotic protein kinases. Proteins 2007; 68 (04) 893-914
  • 28 Omar MH, Scott JD. AKAP signaling islands: venues for precision pharmacology. Trends Pharmacol Sci 2020; 41 (12) 933-946
  • 29 Burgers PP, Ma Y, Margarucci L. et al. A small novel A-kinase anchoring protein (AKAP) that localizes specifically protein kinase A-regulatory subunit I (PKA-RI) to the plasma membrane. J Biol Chem 2012; 287 (52) 43789-43797
  • 30 Raslan Z, Aburima A, Naseem KM. The spatiotemporal regulation of cAMP signaling in blood platelets—old friends and new players. Front Pharmacol 2015; 6: 266
  • 31 Margarucci L, Roest M, Preisinger C. et al. Collagen stimulation of platelets induces a rapid spatial response of cAMP and cGMP signaling scaffolds. Mol Biosyst 2011; 7 (07) 2311-2319
  • 32 Lohmann SM, Vaandrager AB, Smolenski A, Walter U, De Jonge HR. Distinct and specific functions of cGMP-dependent protein kinases. Trends Biochem Sci 1997; 22 (08) 307-312
  • 33 Massberg S, Sausbier M, Klatt P. et al. Increased adhesion and aggregation of platelets lacking cyclic guanosine 3′,5′-monophosphate kinase I. J Exp Med 1999; 189 (08) 1255-1264
  • 34 Gambaryan S, Butt E, Kobsar A. et al. The oligopeptide DT-2 is a specific PKG I inhibitor only in vitro, not in living cells. Br J Pharmacol 2012; 167 (04) 826-838
  • 35 Beck F, Geiger J, Gambaryan S. et al. Time-resolved characterization of cAMP/PKA-dependent signaling reveals that platelet inhibition is a concerted process involving multiple signaling pathways. Blood 2014; 123 (05) e1-e10
  • 36 Beck F, Geiger J, Gambaryan S. et al. Temporal quantitative phosphoproteomics of ADP stimulation reveals novel central nodes in platelet activation and inhibition. Blood 2017; 129 (02) e1-e12
  • 37 Swieringa F, Solari FA, Pagel O. et al. Impaired iloprost-induced platelet inhibition and phosphoproteome changes in patients with confirmed pseudohypoparathyroidism type Ia, linked to genetic mutations in GNAS. Sci Rep 2020; 10 (01) 11389
  • 38 Comer S, Nagy Z, Bolado A. et al. The RhoA regulators Myo9b and GEF-H1 are targets of cyclic nucleotide-dependent kinases in platelets. J Thromb Haemost 2020; 18 (11) 3002-3012
  • 39 Daniel JL, Adelstein RS. Isolation and properties of platelet myosin light chain kinase. Biochemistry 1976; 15 (11) 2370-2377
  • 40 Hathaway DR, Adelstein RS. Human platelet myosin light chain kinase requires the calcium-binding protein calmodulin for activity. Proc Natl Acad Sci U S A 1979; 76 (04) 1653-1657
  • 41 Nishikawa M, de Lanerolle P, Lincoln TM, Adelstein RS. Phosphorylation of mammalian myosin light chain kinases by the catalytic subunit of cyclic AMP-dependent protein kinase and by cyclic GMP-dependent protein kinase. J Biol Chem 1984; 259 (13) 8429-8436
  • 42 Kawahara Y, Takai Y, Minakuchi R, Sano K, Nishizuka Y. Phospholipid turnover as a possible transmembrane signal for protein phosphorylation during human platelet activation by thrombin. Biochem Biophys Res Commun 1980; 97 (01) 309-317
  • 43 Kikkawa U, Nishizuka Y. The role of protein kinase C in transmembrane signalling. Annu Rev Cell Biol 1986; 2: 149-178
  • 44 Kajikawa N, Kikkawa U, Itoh K, Nishizuka Y. Membrane-permeable diacylglycerol, its application to platelet secretion, and regulation of platelet protein kinase C. Methods Enzymol 1989; 169: 430-442
  • 45 Khapchaev AY, Shirinsky VP. Myosin light chain kinase MYLK1: anatomy, interactions, functions, and regulation. Biochemistry (Mosc) 2016; 81 (13) 1676-1697
  • 46 Takai A, Eto M, Hirano K, Takeya K, Wakimoto T, Watanabe M. Protein phosphatases 1 and 2A and their naturally occurring inhibitors: current topics in smooth muscle physiology and chemical biology. J Physiol Sci 2018; 68 (01) 1-17
  • 47 Aburima A, Naseem KM. Platelet myosin light chain phosphatase: keeping it together. Biochem Soc Trans 2014; 42 (02) 279-283
  • 48 Aburima A, Walladbegi K, Wake JD, Naseem KM. cGMP signaling inhibits platelet shape change through regulation of the RhoA-Rho kinase-MLC phosphatase signaling pathway. J Thromb Haemost 2017; 15 (08) 1668-1678
  • 49 Newton AC. Protein kinase C: perfectly balanced. Crit Rev Biochem Mol Biol 2018; 53 (02) 208-230
  • 50 Heemskerk JWM, Harper MT, Cosemans JM, Poole AW. Unravelling the different functions of protein kinase C isoforms in platelets. FEBS Lett 2011; 585 (12) 1711-1716
  • 51 Gilio K, Harper MT, Cosemans JM. et al. Functional divergence of platelet protein kinase C (PKC) isoforms in thrombus formation on collagen. J Biol Chem 2010; 285 (30) 23410-23419
  • 52 Yacoub D, Théorêt JF, Villeneuve L. et al. Essential role of protein kinase C delta in platelet signaling, alpha IIb beta 3 activation, and thromboxane A2 release. J Biol Chem 2006; 281 (40) 30024-30035
  • 53 Mócsai A, Ruland J, Tybulewicz VLJ. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol 2010; 10 (06) 387-402
  • 54 Bradshaw JM. The Src, Syk, and Tec family kinases: distinct types of molecular switches. Cell Signal 2010; 22 (08) 1175-1184
  • 55 Mansueto MS, Reens A, Rakhilina L, Chi A, Pan BS, Miller JR. A reevaluation of the spleen tyrosine kinase (SYK) activation mechanism. J Biol Chem 2019; 294 (19) 7658-7668
  • 56 Grädler U, Schwarz D, Dresing V. et al. Structural and biophysical characterization of the Syk activation switch. J Mol Biol 2013; 425 (02) 309-333
  • 57 Vang T, Torgersen KM, Sundvold V. et al. Activation of the COOH-terminal Src kinase (Csk) by cAMP-dependent protein kinase inhibits signaling through the T cell receptor. J Exp Med 2001; 193 (04) 497-507
  • 58 Abrahamsen H, Vang T, Taskén K. Protein kinase A intersects SRC signaling in membrane microdomains. J Biol Chem 2003; 278 (19) 17170-17177
  • 59 Obergfell A, Eto K, Mocsai A. et al. Coordinate interactions of Csk, Src, and Syk kinases with [alpha]IIb[beta]3 initiate integrin signaling to the cytoskeleton. J Cell Biol 2002; 157 (02) 265-275
  • 60 Schmidt GJ, Reumiller CM, Ercan H. et al. Comparative proteomics reveals unexpected quantitative phosphorylation differences linked to platelet activation state. Sci Rep 2019; 9 (01) 19009
  • 61 Makhoul S, Trabold K, Gambaryan S. et al. cAMP- and cGMP-elevating agents inhibit GPIbα-mediated aggregation but not GPIbα-stimulated Syk activation in human platelets. Cell Commun Signal 2019; 17 (01) 122
  • 62 Makhoul S, Dorschel S, Gambaryan S, Walter U, Jurk K. Feedback regulation of Syk by protein kinase C in human platelets. Int J Mol Sci 2019; 21 (01) 21
  • 63 Makhoul S, Kumm E, Zhang P, Walter U, Jurk K. The serine/threonine protein phosphatase 2A (PP2A) regulates Syk activity in human platelets. Int J Mol Sci 2020; 21 (23) 21
  • 64 Bohnenberger H, Oellerich T, Engelke M, Hsiao HH, Urlaub H, Wienands J. Complex phosphorylation dynamics control the composition of the Syk interactome in B cells. Eur J Immunol 2011; 41 (06) 1550-1562
  • 65 Buffard M, Naldi A, Freiss G. et al. Comparison of SYK signaling networks reveals the potential molecular determinants of its tumor-promoting and suppressing functions. Biomolecules 2021; 11 (02) 308
  • 66 Krisenko MO, Geahlen RL. Calling in SYK: SYK's dual role as a tumor promoter and tumor suppressor in cancer. Biochim Biophys Acta 2015; 1853 (01) 254-263
  • 67 Senis YA. Protein-tyrosine phosphatases: a new frontier in platelet signal transduction. J Thromb Haemost 2013; 11 (10) 1800-1813
  • 68 Tautz L, Senis YA, Oury C, Rahmouni S. Perspective: tyrosine phosphatases as novel targets for antiplatelet therapy. Bioorg Med Chem 2015; 23 (12) 2786-2797
  • 69 Kumm EJ, Pagel O, Gambaryan S. et al. The cell cycle checkpoint system MAST(L)-ENSA/ARPP19-PP2A is targeted by cAMP/PKA and cGMP/PKG in anucleate human platelets. Cells 2020; 9 (02) 9
  • 70 MacDonald JA, Walsh MP. Regulation of smooth muscle myosin light chain phosphatase by multisite phosphorylation of the myosin targeting subunit, MYPT1. Cardiovasc Hematol Disord Drug Targets 2018; 18 (01) 4-13
  • 71 Reynhout S, Janssens V. Physiologic functions of PP2A: lessons from genetically modified mice. Biochim Biophys Acta Mol Cell Res 2019; 1866 (01) 31-50
  • 72 Mochida S, Maslen SL, Skehel M, Hunt T. Greatwall phosphorylates an inhibitor of protein phosphatase 2A that is essential for mitosis. Science 2010; 330 (6011): 1670-1673
  • 73 Gharbi-Ayachi A, Labbé JC, Burgess A. et al. The substrate of Greatwall kinase, Arpp19, controls mitosis by inhibiting protein phosphatase 2A. Science 2010; 330 (6011): 1673-1677
  • 74 Castro A, Lorca T. Greatwall kinase at a glance. J Cell Sci 2018; 131 (20) 131
  • 75 Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2018; 96: 98-134
  • 76 Morita K, He S, Nowak RP. et al. Allosteric activators of protein phosphatase 2A display broad antitumor activity mediated by dephosphorylation of MYBL2. Cell 2020; 181 (03) 702-715.e20
  • 77 Leonard D, Huang W, Izadmehr S. et al. Selective PP2A enhancement through biased heterotrimer stabilization. Cell 2020; 181 (03) 688-701.e16
  • 78 Westermarck J, Neel BG. Piecing together a broken tumor suppressor phosphatase for cancer therapy. Cell 2020; 181 (03) 514-517
  • 79 Yue M, Luo D, Yu S. et al. Misshapen/NIK-related kinase (MINK1) is involved in platelet function, hemostasis, and thrombus formation. Blood 2016; 127 (07) 927-937
  • 80 Fan X, Wang C, Shi P. et al. Platelet MEKK3 regulates arterial thrombosis and myocardial infarct expansion in mice. Blood Adv 2018; 2 (12) 1439-1448
  • 81 Shankar H, Garcia A, Prabhakar J, Kim S, Kunapuli SP. P2Y12 receptor-mediated potentiation of thrombin-induced thromboxane A2 generation in platelets occurs through regulation of Erk1/2 activation. J Thromb Haemost 2006; 4 (03) 638-647
  • 82 Sledz KM, Moore SF, Vijayaragavan V. et al. Redundant role of ASK1-mediated p38MAPK activation in human platelet function. Cell Signal 2020; 68: 109528
  • 83 Naik MU, Patel P, Derstine R. et al. Ask1 regulates murine platelet granule secretion, thromboxane A2 generation, and thrombus formation. Blood 2017; 129 (09) 1197-1209
  • 84 Kamiyama M, Shirai T, Tamura S. et al. ASK1 facilitates tumor metastasis through phosphorylation of an ADP receptor P2Y12 in platelets. Cell Death Differ 2017; 24 (12) 2066-2076
  • 85 Liu Q, Hofmann PA. Protein phosphatase 2A-mediated cross-talk between p38 MAPK and ERK in apoptosis of cardiac myocytes. Am J Physiol Heart Circ Physiol 2004; 286 (06) H2204-H2212
  • 86 Tullemans BME, Heemskerk JWM, Kuijpers MJE. Acquired platelet antagonism: off-target antiplatelet effects of malignancy treatment with tyrosine kinase inhibitors. J Thromb Haemost 2018; 16 (09) 1686-1699
  • 87 Busygina K, Denzinger V, Bernlochner I, Weber C, Lorenz R, Siess W. Btk inhibitors as first oral atherothrombosis-selective antiplatelet drugs?. Thromb Haemost 2019; 119 (08) 1212-1221
  • 88 Series J, Ribes A, Garcia C. et al. Effects of novel Btk and Syk inhibitors on platelet functions alone and in combination in vitro and in vivo. J Thromb Haemost 2020; 18 (12) 3336-3351
  • 89 Sangodkar J, Farrington CC, McClinch K, Galsky MD, Kastrinsky DB, Narla G. All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J 2016; 283 (06) 1004-1024
  • 90 O'Connor CM, Perl A, Leonard D, Sangodkar J, Narla G. Therapeutic targeting of PP2A. Int J Biochem Cell Biol 2018; 96: 182-193
  • 91 Séverin S, Ghevaert C, Mazharian A. The mitogen-activated protein kinase signaling pathways: role in megakaryocyte differentiation. J Thromb Haemost 2010; 8 (01) 17-26
  • 92 Di Buduo CA, Currao M, Pecci A, Kaplan DL, Balduini CL, Balduini A. Revealing eltrombopag's promotion of human megakaryopoiesis through AKT/ERK-dependent pathway activation. Haematologica 2016; 101 (12) 1479-1488
  • 93 Bluteau D, Balduini A, Balayn N. et al. Thrombocytopenia-associated mutations in the ANKRD26 regulatory region induce MAPK hyperactivation. J Clin Invest 2014; 124 (02) 580-591
  • 94 Desterke C, Bilhou-Nabéra C, Guerton B. et al; French Intergroup of Myeloproliferative Disorders, French INSERM, European EUMNET Networks on Myelofibrosis. FLT3-mediated p38-MAPK activation participates in the control of megakaryopoiesis in primary myelofibrosis. Cancer Res 2011; 71 (08) 2901-2915
  • 95 Hurtado B, Trakala M, Ximénez-Embún P. et al. Thrombocytopenia-associated mutations in Ser/Thr kinase MASTL deregulate actin cytoskeletal dynamics in platelets. J Clin Invest 2018; 128 (12) 5351-5367
  • 96 Bekker-Jensen DB, Bernhardt OM, Hogrebe A. et al. Rapid and site-specific deep phosphoproteome profiling by data-independent acquisition without the need for spectral libraries. Nat Commun 2020; 11 (01) 787
  • 97 Roskoski Jr R. Properties of FDA-approved small molecule protein kinase inhibitors: a 2020 update. Pharmacol Res 2020; 152: 104609
  • 98 Babur Ö, Melrose AR, Cunliffe JM. et al. Phosphoproteomic quantitation and causal analysis reveal pathways in GPVI/ITAM-mediated platelet activation programs. Blood 2020; 136 (20) 2346-2358
  • 99 Aslan JE. Platelet proteomes, pathways, and phenotypes as informants of vascular wellness and disease. Arterioscler Thromb Vasc Biol 2021; 41 (03) 999-1011
  • 100 Stefanini L, Lee RH, Bergmeier W. GTPases. In: Platelets in Thrombotic and Non-Thrombotic Disorders: Pathophysiology, Pharmacology and Therapeutics: An Update. Springer International Publishing; 2017: 263-284
  • 101 Lee RH, Stefanini L, Bergmeier W. Platelet signal transduction. In: Platelets. 4th ed.. Academic Press; 2019: 329-348
  • 102 Hubertus K, Mischnik M, Timmer J. et al. Reciprocal regulation of human platelet function by endogenous prostanoids and through multiple prostanoid receptors. Eur J Pharmacol 2014; 740: 15-27
  • 103 Yeung J, Li W, Holinstat M. Platelet signaling and disease: targeted therapy for thrombosis and other related diseases. Pharmacol Rev 2018; 70 (03) 526-548
  • 104 Chatterjee M, Rath D, Gawaz M. Role of chemokine receptors CXCR4 and CXCR7 for platelet function. Biochem Soc Trans 2015; 43 (04) 720-726
  • 105 Cheng Z, Gao W, Fan X. et al. Extracellular signal-regulated kinase 5 associates with casein kinase II to regulate GPIb-IX-mediated platelet activation via the PTEN/PI3K/Akt pathway. J Thromb Haemost 2017; 15 (08) 1679-1688
  • 106 Yang M, Kholmukhamedov A, Schulte ML. et al. Platelet CD36 signaling through ERK5 promotes caspase-dependent procoagulant activity and fibrin deposition in vivo. Blood Adv 2018; 2 (21) 2848-2861
  • 107 Manikanta K, Naveen Kumar SK, Hemshekhar M, Kemparaju K, Girish KS. ASK1 inhibition triggers platelet apoptosis via p38-MAPK-mediated mitochondrial dysfunction. Haematologica 2020; 105 (08) e419-e423

Address for correspondence

Kerstin Jurk, PhD
Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz
Langenbeckstr. 1, 55131 Mainz
Germany   

Publication History

Received: 23 December 2020

Accepted: 06 April 2021

Article published online:
30 June 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Born GV. Aggregation of blood platelets by adenosine diphosphate and its reversal. Nature 1962; 194: 927-929
  • 2 Svensson J, Hamberg M, Samuelsson B. On the formation and effects of thromboxane A2 in human platelets. Acta Physiol Scand 1976; 98 (03) 285-294
  • 3 Moncada S, Higgs EA, Vane JR. Human arterial and venous tissues generate prostacyclin (prostaglandin x), a potent inhibitor of platelet aggregation. Lancet 1977; 1 (8001): 18-20
  • 4 Tateson JE, Moncada S, Vane JR. Effects of prostacyclin (PGX) on cyclic AMP concentrations in human platelets. Prostaglandins 1977; 13 (03) 389-397
  • 5 Jurk K, Kehrel BE. Platelets: physiology and biochemistry. Semin Thromb Hemost 2005; 31 (04) 381-392
  • 6 Versteeg HH, Heemskerk JWM, Levi M, Reitsma PH. New fundamentals in hemostasis. Physiol Rev 2013; 93 (01) 327-358
  • 7 Jurk K. Analysis of platelet function and dysfunction. Hamostaseologie 2015; 35 (01) 60-72
  • 8 Senis YA, Mazharian A, Mori J. Src family kinases: at the forefront of platelet activation. Blood 2014; 124 (13) 2013-2024
  • 9 Brass LF, Ma P, Tomaiuolo M. et al. A systems approach to the platelet signaling network and the hemostatic response to injury. In: Platelets in Thrombotic and Non-Thrombotic Disorders. Springer; 2017: 367-378
  • 10 Makhoul S, Walter E, Pagel O. et al. Effects of the NO/soluble guanylate cyclase/cGMP system on the functions of human platelets. Nitric Oxide 2018; 76: 71-80
  • 11 Sutherland EW, Robison GA. The role of cyclic-3′,5′-AMP in responses to catecholamines and other hormones. Pharmacol Rev 1966; 18 (01) 145-161
  • 12 Sutherland EW. On the biological role of cyclic AMP. JAMA 1970; 214 (07) 1281-1288
  • 13 Mellion BT, Ignarro LJ, Ohlstein EH, Pontecorvo EG, Hyman AL, Kadowitz PJ. Evidence for the inhibitory role of guanosine 3′, 5′-monophosphate in ADP-induced human platelet aggregation in the presence of nitric oxide and related vasodilators. Blood 1981; 57 (05) 946-955
  • 14 Eigenthaler M, Ullrich H, Geiger J. et al. Defective nitrovasodilator-stimulated protein phosphorylation and calcium regulation in cGMP-dependent protein kinase-deficient human platelets of chronic myelocytic leukemia. J Biol Chem 1993; 268 (18) 13526-13531
  • 15 Waldmann R, Bauer S, Göbel C, Hofmann F, Jakobs KH, Walter U. Demonstration of cGMP-dependent protein kinase and cGMP-dependent phosphorylation in cell-free extracts of platelets. Eur J Biochem 1986; 158 (01) 203-210
  • 16 Smolenski A. Novel roles of cAMP/cGMP-dependent signaling in platelets. J Thromb Haemost 2012; 10 (02) 167-176
  • 17 Nagy Z, Smolenski A. Cyclic nucleotide-dependent inhibitory signaling interweaves with activating pathways to determine platelet responses. Res Pract Thromb Haemost 2018; 2 (03) 558-571
  • 18 Kunapuli SP, Bhavanasi D, Kostyak JC. et al. Platelet signaling: protein phosphorylation. In: Platelets in Thrombotic and Non-Thrombotic Disorders: Pathophysiology, Pharmacology and Therapeutics: An Update. Springer International Publishing; 2017: 297-308
  • 19 Golden A, Nemeth SP, Brugge JS. Blood platelets express high levels of the pp60c-src-specific tyrosine kinase activity. Proc Natl Acad Sci U S A 1986; 83 (04) 852-856
  • 20 Séverin S, Nash CA, Mori J. et al. Distinct and overlapping functional roles of Src family kinases in mouse platelets. J Thromb Haemost 2012; 10 (08) 1631-1645
  • 21 Patel P, Naik UP. Platelet MAPKs-a 20+ year history: What do we really know?. J Thromb Haemost 2020; 18 (09) 2087-2102
  • 22 Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science 2002; 298 (5600): 1912-1934
  • 23 Chen MJ, Dixon JE, Manning G. Genomics and evolution of protein phosphatases. Sci Signal 2017; 10 (474) 10
  • 24 Burkhart JM, Vaudel M, Gambaryan S. et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 2012; 120 (15) e73-e82
  • 25 Zeiler M, Moser M, Mann M. Copy number analysis of the murine platelet proteome spanning the complete abundance range. Mol Cell Proteomics 2014; 13 (12) 3435-3445
  • 26 Jurk K, Walter U. New Insights into platelet signalling pathways by functional and proteomic approaches. Hamostaseologie 2019; 39 (02) 140-151
  • 27 Miranda-Saavedra D, Barton GJ. Classification and functional annotation of eukaryotic protein kinases. Proteins 2007; 68 (04) 893-914
  • 28 Omar MH, Scott JD. AKAP signaling islands: venues for precision pharmacology. Trends Pharmacol Sci 2020; 41 (12) 933-946
  • 29 Burgers PP, Ma Y, Margarucci L. et al. A small novel A-kinase anchoring protein (AKAP) that localizes specifically protein kinase A-regulatory subunit I (PKA-RI) to the plasma membrane. J Biol Chem 2012; 287 (52) 43789-43797
  • 30 Raslan Z, Aburima A, Naseem KM. The spatiotemporal regulation of cAMP signaling in blood platelets—old friends and new players. Front Pharmacol 2015; 6: 266
  • 31 Margarucci L, Roest M, Preisinger C. et al. Collagen stimulation of platelets induces a rapid spatial response of cAMP and cGMP signaling scaffolds. Mol Biosyst 2011; 7 (07) 2311-2319
  • 32 Lohmann SM, Vaandrager AB, Smolenski A, Walter U, De Jonge HR. Distinct and specific functions of cGMP-dependent protein kinases. Trends Biochem Sci 1997; 22 (08) 307-312
  • 33 Massberg S, Sausbier M, Klatt P. et al. Increased adhesion and aggregation of platelets lacking cyclic guanosine 3′,5′-monophosphate kinase I. J Exp Med 1999; 189 (08) 1255-1264
  • 34 Gambaryan S, Butt E, Kobsar A. et al. The oligopeptide DT-2 is a specific PKG I inhibitor only in vitro, not in living cells. Br J Pharmacol 2012; 167 (04) 826-838
  • 35 Beck F, Geiger J, Gambaryan S. et al. Time-resolved characterization of cAMP/PKA-dependent signaling reveals that platelet inhibition is a concerted process involving multiple signaling pathways. Blood 2014; 123 (05) e1-e10
  • 36 Beck F, Geiger J, Gambaryan S. et al. Temporal quantitative phosphoproteomics of ADP stimulation reveals novel central nodes in platelet activation and inhibition. Blood 2017; 129 (02) e1-e12
  • 37 Swieringa F, Solari FA, Pagel O. et al. Impaired iloprost-induced platelet inhibition and phosphoproteome changes in patients with confirmed pseudohypoparathyroidism type Ia, linked to genetic mutations in GNAS. Sci Rep 2020; 10 (01) 11389
  • 38 Comer S, Nagy Z, Bolado A. et al. The RhoA regulators Myo9b and GEF-H1 are targets of cyclic nucleotide-dependent kinases in platelets. J Thromb Haemost 2020; 18 (11) 3002-3012
  • 39 Daniel JL, Adelstein RS. Isolation and properties of platelet myosin light chain kinase. Biochemistry 1976; 15 (11) 2370-2377
  • 40 Hathaway DR, Adelstein RS. Human platelet myosin light chain kinase requires the calcium-binding protein calmodulin for activity. Proc Natl Acad Sci U S A 1979; 76 (04) 1653-1657
  • 41 Nishikawa M, de Lanerolle P, Lincoln TM, Adelstein RS. Phosphorylation of mammalian myosin light chain kinases by the catalytic subunit of cyclic AMP-dependent protein kinase and by cyclic GMP-dependent protein kinase. J Biol Chem 1984; 259 (13) 8429-8436
  • 42 Kawahara Y, Takai Y, Minakuchi R, Sano K, Nishizuka Y. Phospholipid turnover as a possible transmembrane signal for protein phosphorylation during human platelet activation by thrombin. Biochem Biophys Res Commun 1980; 97 (01) 309-317
  • 43 Kikkawa U, Nishizuka Y. The role of protein kinase C in transmembrane signalling. Annu Rev Cell Biol 1986; 2: 149-178
  • 44 Kajikawa N, Kikkawa U, Itoh K, Nishizuka Y. Membrane-permeable diacylglycerol, its application to platelet secretion, and regulation of platelet protein kinase C. Methods Enzymol 1989; 169: 430-442
  • 45 Khapchaev AY, Shirinsky VP. Myosin light chain kinase MYLK1: anatomy, interactions, functions, and regulation. Biochemistry (Mosc) 2016; 81 (13) 1676-1697
  • 46 Takai A, Eto M, Hirano K, Takeya K, Wakimoto T, Watanabe M. Protein phosphatases 1 and 2A and their naturally occurring inhibitors: current topics in smooth muscle physiology and chemical biology. J Physiol Sci 2018; 68 (01) 1-17
  • 47 Aburima A, Naseem KM. Platelet myosin light chain phosphatase: keeping it together. Biochem Soc Trans 2014; 42 (02) 279-283
  • 48 Aburima A, Walladbegi K, Wake JD, Naseem KM. cGMP signaling inhibits platelet shape change through regulation of the RhoA-Rho kinase-MLC phosphatase signaling pathway. J Thromb Haemost 2017; 15 (08) 1668-1678
  • 49 Newton AC. Protein kinase C: perfectly balanced. Crit Rev Biochem Mol Biol 2018; 53 (02) 208-230
  • 50 Heemskerk JWM, Harper MT, Cosemans JM, Poole AW. Unravelling the different functions of protein kinase C isoforms in platelets. FEBS Lett 2011; 585 (12) 1711-1716
  • 51 Gilio K, Harper MT, Cosemans JM. et al. Functional divergence of platelet protein kinase C (PKC) isoforms in thrombus formation on collagen. J Biol Chem 2010; 285 (30) 23410-23419
  • 52 Yacoub D, Théorêt JF, Villeneuve L. et al. Essential role of protein kinase C delta in platelet signaling, alpha IIb beta 3 activation, and thromboxane A2 release. J Biol Chem 2006; 281 (40) 30024-30035
  • 53 Mócsai A, Ruland J, Tybulewicz VLJ. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol 2010; 10 (06) 387-402
  • 54 Bradshaw JM. The Src, Syk, and Tec family kinases: distinct types of molecular switches. Cell Signal 2010; 22 (08) 1175-1184
  • 55 Mansueto MS, Reens A, Rakhilina L, Chi A, Pan BS, Miller JR. A reevaluation of the spleen tyrosine kinase (SYK) activation mechanism. J Biol Chem 2019; 294 (19) 7658-7668
  • 56 Grädler U, Schwarz D, Dresing V. et al. Structural and biophysical characterization of the Syk activation switch. J Mol Biol 2013; 425 (02) 309-333
  • 57 Vang T, Torgersen KM, Sundvold V. et al. Activation of the COOH-terminal Src kinase (Csk) by cAMP-dependent protein kinase inhibits signaling through the T cell receptor. J Exp Med 2001; 193 (04) 497-507
  • 58 Abrahamsen H, Vang T, Taskén K. Protein kinase A intersects SRC signaling in membrane microdomains. J Biol Chem 2003; 278 (19) 17170-17177
  • 59 Obergfell A, Eto K, Mocsai A. et al. Coordinate interactions of Csk, Src, and Syk kinases with [alpha]IIb[beta]3 initiate integrin signaling to the cytoskeleton. J Cell Biol 2002; 157 (02) 265-275
  • 60 Schmidt GJ, Reumiller CM, Ercan H. et al. Comparative proteomics reveals unexpected quantitative phosphorylation differences linked to platelet activation state. Sci Rep 2019; 9 (01) 19009
  • 61 Makhoul S, Trabold K, Gambaryan S. et al. cAMP- and cGMP-elevating agents inhibit GPIbα-mediated aggregation but not GPIbα-stimulated Syk activation in human platelets. Cell Commun Signal 2019; 17 (01) 122
  • 62 Makhoul S, Dorschel S, Gambaryan S, Walter U, Jurk K. Feedback regulation of Syk by protein kinase C in human platelets. Int J Mol Sci 2019; 21 (01) 21
  • 63 Makhoul S, Kumm E, Zhang P, Walter U, Jurk K. The serine/threonine protein phosphatase 2A (PP2A) regulates Syk activity in human platelets. Int J Mol Sci 2020; 21 (23) 21
  • 64 Bohnenberger H, Oellerich T, Engelke M, Hsiao HH, Urlaub H, Wienands J. Complex phosphorylation dynamics control the composition of the Syk interactome in B cells. Eur J Immunol 2011; 41 (06) 1550-1562
  • 65 Buffard M, Naldi A, Freiss G. et al. Comparison of SYK signaling networks reveals the potential molecular determinants of its tumor-promoting and suppressing functions. Biomolecules 2021; 11 (02) 308
  • 66 Krisenko MO, Geahlen RL. Calling in SYK: SYK's dual role as a tumor promoter and tumor suppressor in cancer. Biochim Biophys Acta 2015; 1853 (01) 254-263
  • 67 Senis YA. Protein-tyrosine phosphatases: a new frontier in platelet signal transduction. J Thromb Haemost 2013; 11 (10) 1800-1813
  • 68 Tautz L, Senis YA, Oury C, Rahmouni S. Perspective: tyrosine phosphatases as novel targets for antiplatelet therapy. Bioorg Med Chem 2015; 23 (12) 2786-2797
  • 69 Kumm EJ, Pagel O, Gambaryan S. et al. The cell cycle checkpoint system MAST(L)-ENSA/ARPP19-PP2A is targeted by cAMP/PKA and cGMP/PKG in anucleate human platelets. Cells 2020; 9 (02) 9
  • 70 MacDonald JA, Walsh MP. Regulation of smooth muscle myosin light chain phosphatase by multisite phosphorylation of the myosin targeting subunit, MYPT1. Cardiovasc Hematol Disord Drug Targets 2018; 18 (01) 4-13
  • 71 Reynhout S, Janssens V. Physiologic functions of PP2A: lessons from genetically modified mice. Biochim Biophys Acta Mol Cell Res 2019; 1866 (01) 31-50
  • 72 Mochida S, Maslen SL, Skehel M, Hunt T. Greatwall phosphorylates an inhibitor of protein phosphatase 2A that is essential for mitosis. Science 2010; 330 (6011): 1670-1673
  • 73 Gharbi-Ayachi A, Labbé JC, Burgess A. et al. The substrate of Greatwall kinase, Arpp19, controls mitosis by inhibiting protein phosphatase 2A. Science 2010; 330 (6011): 1673-1677
  • 74 Castro A, Lorca T. Greatwall kinase at a glance. J Cell Sci 2018; 131 (20) 131
  • 75 Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2018; 96: 98-134
  • 76 Morita K, He S, Nowak RP. et al. Allosteric activators of protein phosphatase 2A display broad antitumor activity mediated by dephosphorylation of MYBL2. Cell 2020; 181 (03) 702-715.e20
  • 77 Leonard D, Huang W, Izadmehr S. et al. Selective PP2A enhancement through biased heterotrimer stabilization. Cell 2020; 181 (03) 688-701.e16
  • 78 Westermarck J, Neel BG. Piecing together a broken tumor suppressor phosphatase for cancer therapy. Cell 2020; 181 (03) 514-517
  • 79 Yue M, Luo D, Yu S. et al. Misshapen/NIK-related kinase (MINK1) is involved in platelet function, hemostasis, and thrombus formation. Blood 2016; 127 (07) 927-937
  • 80 Fan X, Wang C, Shi P. et al. Platelet MEKK3 regulates arterial thrombosis and myocardial infarct expansion in mice. Blood Adv 2018; 2 (12) 1439-1448
  • 81 Shankar H, Garcia A, Prabhakar J, Kim S, Kunapuli SP. P2Y12 receptor-mediated potentiation of thrombin-induced thromboxane A2 generation in platelets occurs through regulation of Erk1/2 activation. J Thromb Haemost 2006; 4 (03) 638-647
  • 82 Sledz KM, Moore SF, Vijayaragavan V. et al. Redundant role of ASK1-mediated p38MAPK activation in human platelet function. Cell Signal 2020; 68: 109528
  • 83 Naik MU, Patel P, Derstine R. et al. Ask1 regulates murine platelet granule secretion, thromboxane A2 generation, and thrombus formation. Blood 2017; 129 (09) 1197-1209
  • 84 Kamiyama M, Shirai T, Tamura S. et al. ASK1 facilitates tumor metastasis through phosphorylation of an ADP receptor P2Y12 in platelets. Cell Death Differ 2017; 24 (12) 2066-2076
  • 85 Liu Q, Hofmann PA. Protein phosphatase 2A-mediated cross-talk between p38 MAPK and ERK in apoptosis of cardiac myocytes. Am J Physiol Heart Circ Physiol 2004; 286 (06) H2204-H2212
  • 86 Tullemans BME, Heemskerk JWM, Kuijpers MJE. Acquired platelet antagonism: off-target antiplatelet effects of malignancy treatment with tyrosine kinase inhibitors. J Thromb Haemost 2018; 16 (09) 1686-1699
  • 87 Busygina K, Denzinger V, Bernlochner I, Weber C, Lorenz R, Siess W. Btk inhibitors as first oral atherothrombosis-selective antiplatelet drugs?. Thromb Haemost 2019; 119 (08) 1212-1221
  • 88 Series J, Ribes A, Garcia C. et al. Effects of novel Btk and Syk inhibitors on platelet functions alone and in combination in vitro and in vivo. J Thromb Haemost 2020; 18 (12) 3336-3351
  • 89 Sangodkar J, Farrington CC, McClinch K, Galsky MD, Kastrinsky DB, Narla G. All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J 2016; 283 (06) 1004-1024
  • 90 O'Connor CM, Perl A, Leonard D, Sangodkar J, Narla G. Therapeutic targeting of PP2A. Int J Biochem Cell Biol 2018; 96: 182-193
  • 91 Séverin S, Ghevaert C, Mazharian A. The mitogen-activated protein kinase signaling pathways: role in megakaryocyte differentiation. J Thromb Haemost 2010; 8 (01) 17-26
  • 92 Di Buduo CA, Currao M, Pecci A, Kaplan DL, Balduini CL, Balduini A. Revealing eltrombopag's promotion of human megakaryopoiesis through AKT/ERK-dependent pathway activation. Haematologica 2016; 101 (12) 1479-1488
  • 93 Bluteau D, Balduini A, Balayn N. et al. Thrombocytopenia-associated mutations in the ANKRD26 regulatory region induce MAPK hyperactivation. J Clin Invest 2014; 124 (02) 580-591
  • 94 Desterke C, Bilhou-Nabéra C, Guerton B. et al; French Intergroup of Myeloproliferative Disorders, French INSERM, European EUMNET Networks on Myelofibrosis. FLT3-mediated p38-MAPK activation participates in the control of megakaryopoiesis in primary myelofibrosis. Cancer Res 2011; 71 (08) 2901-2915
  • 95 Hurtado B, Trakala M, Ximénez-Embún P. et al. Thrombocytopenia-associated mutations in Ser/Thr kinase MASTL deregulate actin cytoskeletal dynamics in platelets. J Clin Invest 2018; 128 (12) 5351-5367
  • 96 Bekker-Jensen DB, Bernhardt OM, Hogrebe A. et al. Rapid and site-specific deep phosphoproteome profiling by data-independent acquisition without the need for spectral libraries. Nat Commun 2020; 11 (01) 787
  • 97 Roskoski Jr R. Properties of FDA-approved small molecule protein kinase inhibitors: a 2020 update. Pharmacol Res 2020; 152: 104609
  • 98 Babur Ö, Melrose AR, Cunliffe JM. et al. Phosphoproteomic quantitation and causal analysis reveal pathways in GPVI/ITAM-mediated platelet activation programs. Blood 2020; 136 (20) 2346-2358
  • 99 Aslan JE. Platelet proteomes, pathways, and phenotypes as informants of vascular wellness and disease. Arterioscler Thromb Vasc Biol 2021; 41 (03) 999-1011
  • 100 Stefanini L, Lee RH, Bergmeier W. GTPases. In: Platelets in Thrombotic and Non-Thrombotic Disorders: Pathophysiology, Pharmacology and Therapeutics: An Update. Springer International Publishing; 2017: 263-284
  • 101 Lee RH, Stefanini L, Bergmeier W. Platelet signal transduction. In: Platelets. 4th ed.. Academic Press; 2019: 329-348
  • 102 Hubertus K, Mischnik M, Timmer J. et al. Reciprocal regulation of human platelet function by endogenous prostanoids and through multiple prostanoid receptors. Eur J Pharmacol 2014; 740: 15-27
  • 103 Yeung J, Li W, Holinstat M. Platelet signaling and disease: targeted therapy for thrombosis and other related diseases. Pharmacol Rev 2018; 70 (03) 526-548
  • 104 Chatterjee M, Rath D, Gawaz M. Role of chemokine receptors CXCR4 and CXCR7 for platelet function. Biochem Soc Trans 2015; 43 (04) 720-726
  • 105 Cheng Z, Gao W, Fan X. et al. Extracellular signal-regulated kinase 5 associates with casein kinase II to regulate GPIb-IX-mediated platelet activation via the PTEN/PI3K/Akt pathway. J Thromb Haemost 2017; 15 (08) 1679-1688
  • 106 Yang M, Kholmukhamedov A, Schulte ML. et al. Platelet CD36 signaling through ERK5 promotes caspase-dependent procoagulant activity and fibrin deposition in vivo. Blood Adv 2018; 2 (21) 2848-2861
  • 107 Manikanta K, Naveen Kumar SK, Hemshekhar M, Kemparaju K, Girish KS. ASK1 inhibition triggers platelet apoptosis via p38-MAPK-mediated mitochondrial dysfunction. Haematologica 2020; 105 (08) e419-e423

Zoom Image
Fig. 1 Multiple intracellular serine/threonine protein kinases mediate the functional responses of platelet membrane receptor stimulation and subsequent second messenger signaling. Numerous physiological and pharmacological platelet activators or inhibitors achieve their effects via specific membrane receptors. G-protein–coupled receptors (GPCRs) activate distinct G-proteins (Gq, Gi/z, Gs), phospholipases C (PLC), and their specific second messenger systems (Ca2+, DAG, PIP3, cAMP) and protein kinase targets. Platelet “pattern-recognition” receptors activate Src family tyrosine kinases (SFK)/immunoreceptor tyrosine-based activation motif (ITAM)/spleen tyrosine kinase (Syk) signaling complexes, which also lead to elevated cytoplasmic levels of second messengers and platelet activation. NO, an endothelium-derived gaseous platelet inhibitor, passes through the platelet membrane, stimulates the soluble guanylyl cyclase (sGC), and increases cytoplasmic cGMP levels. Both the cAMP/PKA and cGMP/PKG systems represent potent inhibitory pathways, which block agonist-induced platelet activation at several steps due to the phosphorylation of multiple substrates. The second messengers Ca2+, DAG, and PIP3 also stimulate specific protein kinases, leading to partially overlapping platelet responses. Similar to PKA/PKG, most of these protein kinases also have multiple substrates, which have been only partially identified. This model shown here and many others published in the field have important limitations as discussed in the text. Platelets actually have hundreds of serine/threonine protein kinases (see [Supplementary Table S1] [online only]) with thousands of substrates and interact with other components such as protein phosphatases to integrate the signaling response to a functional response. This not only occurs at a vertical (receptor-to-function) level, but also at horizontal level of signaling (interaction between various protein kinases). We are only at the beginning to understand this very dynamic and rapidly responding protein kinase network, which is addressed in this review. This model is based on previous models published.[6] [100] [101] [102] [103] [104] 5-HT2A, serotonin receptor; CLEC-2, C-type lectin like-receptor 2; CXCR4/CXCR6/CXCR7, chemokine receptors; DP1, prostanoid receptor D1; EP 2,4, prostanoid receptors EP 2, 4; EP1, EP3, prostanoid receptors EP1 and EP3; GPIb–V–IX, glycoprotein Ib–V–IX complex; GPVI, glycoprotein VI; IP, prostanoid/prostacyclin receptor I; P2Y1, ADP/P2 purinoceptor subtype Y1; P2Y12, ADP/P2 purinoceptor subtype Y12; PAR 1/4, protease-activated receptor 1 and 4; TP, thromboxane receptor; α2A, α2A-adrenergic receptor.
Zoom Image
Fig. 2 Model showing activation, inhibition, and fine-tuning of adhesion-dependent Syk regulation in human platelets. Activated GPVI and GPIbα increase SFK-mediated dual phosphorylation of FcRγ ITAM motifs (ppITAM), which recruit inactive Syk (via its SH2 domains) from the cytosol to the cell membrane. This induces a conformational change leading to Syk activation and concomitant Y-phosphorylation of the interdomain B (Y352) and kinase domain (Y525/6), mainly by SFKs and autophosphorylation, respectively. Fully activated Syk and Syk interacting proteins such as LAT, Btk, PLCγ2, E3 ubiquitin-protein ligase CBL form a signalosome complex to trigger various cellular responses. Often, Syk tyrosine phosphorylates its interacting proteins in such complexes. GPVI ligands such as collagen, VWF, and convulxin also induce a Syk- and PKC-mediated S-phosphorylation (S297) within the interdomain B of Syk, which is negatively associated with Y-phosphorylation (Y352, Y525, Y526). Inhibition of the serine/threonine protein phosphatase PP2A by OA also results in Syk S297 phosphorylation due to inhibited dephosphorylation, introducing PP2A as novel factor for Syk regulation. PKA inhibits PKC-mediated Syk S297 phosphorylation at the level of generation and/or action of the secondary mediators ADP and TxA2. The tyrosine phosphatase TULA-2 downregulates Syk tyrosine phosphorylation (mainly pY352) and activity. Syk and its interdomain B are “signaling nodes” for the regulation by multiple serine/threonine and tyrosine protein kinases and phosphatases. ADP, adenosine diphosphate; Btk, Bruton tyrosine kinase; cAMP, cyclic adenosine monophosphate; Cbl, casitas B-lineage lymphoma; cGMP, cyclic guanosine monophosphate; DAG, diacylglycerol; FcRγ, Fc receptor γ-chain; GPCRs, G-protein–coupled receptors; GPVI, glycoprotein VI; IP3, inositol trisphosphate; LAT, linker for activation of T-cells; OA, okadaic acid; PI3K, phosphatidylinositol 3-kinase; PKA, protein kinase A; PKC, protein kinase C; PLCγ2, phospholipase Cγ2; PP2A, protein phosphatase 2A; SFK, src kinase family; SLP-76, Src homology (SH) 2 domain-containing leukocyte phosphoprotein of 76 kDa; Syk, spleen tyrosine kinase; TULA-2, T-cell ubiquitin ligand-2; TxA2, thromboxane A2; VWF, von Willebrand factor.
Zoom Image
Fig. 3 Signaling network of MAPK in human and murine platelets. Black and red lines demonstrate common activatory and inhibitory signaling pathways in human and murine platelets, respectively. Mouse- and human-specific signaling pathways are shown in blue and gray, respectively. (Modified from various studies.[21] [79] [82] [105] [106] [107]) AA, arachidonic acid; ADP, adenosine diphosphate; APP, amyloid precursor protein; ASK1 (MAP3K5), apoptosis signal-regulating kinase 1; JNK1/2, c-Jun N-terminal kinase 1/2; p38 (MAPK14), mitogen-activated protein kinase 14; cPLA2, cytosolic phospholipases A2; ERK 1/2 (MAPK3/MAPK1), mitogen-activated protein kinase 3 and 1, extracellular signal-regulated kinase 1 and 2; FcRγ, Fc receptor γ-chain; JNK1/2 (MAPK 8 and 9), mitogen-activated protein kinase 8 and 9; MAP3K3, mitogen-activated protein kinase kinase kinase 3; MEK (MAP2K) 1/2, 5, MAP kinase kinases 1/2 and 5; MINK1 (MAP4K6), Misshapen-like kinase 1; MK2, MAPK-activated protein kinase 2; MKK (MAP2K), MAP kinase kinases; NLRP3, NLR family pyrin domain containing 3; NOX, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase; oxLDL, oxidized low-density lipoprotein; PARs, protease-activated receptors; PKC, protein kinase C; PP2A, protein phosphatase 2A; RAF, rapidly accelerated fibrosarcoma; ROS, reactive oxygen species; Syk, spleen tyrosine kinase; TP, thromboxane receptor; TxA2 receptor; TxA2, thromboxane A2; VWF, von Willebrand factor.