The importance of the second messengers calcium (Ca2+) and diacylglycerol (DAG) in platelet signal transduction was established more than 30 years ago. Whereas protein kinase C (PKC) family members were discovered as the targets of DAG, little is known about the molecular identity of the main Ca2+ sensor(s). We here identify Ca2+ and DAG-regulated guanine nucleotide exchange factor I (CalDAG-GEFI) as a critical molecule in Ca2+-dependent platelet activation. CalDAG-GEFI, through activation of the small GTPase Rap1, directly triggers integrin activation and extracellular signal-regulated kinase-dependent thromboxane A2 (TxA2) release. CalDAG-GEFI–dependent TxA2 generation provides crucial feedback for PKC activation and granule release, particularly at threshold agonist concentrations. PKC/P2Y12 signaling in turn mediates a second wave of Rap1 activation, necessary for sustained platelet activation and thrombus stabilization. Our results lead to a revised model for platelet activation that establishes one molecule, CalDAG-GEFI, at the nexus of Ca2+-induced integrin activation, TxA2 generation, and granule release. The preferential activation of CalDAG-GEFI over PKC downstream of phospholipase C activation, and the different kinetics of CalDAG-GEFI– and PKC/P2Y12-mediated Rap1 activation demonstrate an unexpected complexity to the platelet activation process, and they challenge the current model that DAG/PKC-dependent signaling events are crucial for the initiation of platelet adhesion.

Platelet thrombus formation at sites of vascular injury is a complex and dynamic process that occurs in several phases.1,2  Platelet preactivation and tethering (transient adhesion) are mediated by the interaction of glycoprotein (GP) lbα with matrix-bound von Willebrand factor, particularly in conditions of high shear. Platelet activation and firm adhesion are then triggered by collagen exposed in the vessel wall and/or locally generated thrombin. Sustained integrin activation and the formation of stable thrombi further depend on persistent costimulatory signaling provided by the second wave agonists, thromboxane A2 (TxA2) and adenosine 5′-diphosphate (ADP), which are released from activated platelets.3-5 

The central biochemical event in platelet aggregation is the agonist-induced inside-out activation of αIIbβ3 integrin. Most platelet agonists initiate intracellular signaling through the activation of phospholipase C (PLC), followed by the generation of the second messengers calcium (Ca2+) and diacylgycerol (DAG). DAG is critical for protein kinase C (PKC) activation, a key event in platelet granule release and integrin activation. Platelets express at least 6 PKC isoforms, which have activating as well as inhibitory roles in platelet activation.6-10  Early studies with calcium ionophores established a role of Ca2+ in integrin activation, TxA2 generation, and granule release.11-13  Initially, Ca2+-binding classical isoforms of PKC were suggested as the cell's main signal integrators for elevated intracellular Ca2+ concentrations ([Ca2+]i). However, more recent studies using calcium chelators in combination with broad range PKC inhibitors identified an independent Ca2+-sensitive signaling pathway that acts synergistically with PKC in the activation of integrin αIIbβ3.14,15  Whereas the role of Ca2+ and DAG/PKC in various aspects of platelet activation has been well documented, the molecular nature of the Ca2+ sensor(s) and the interplay between Ca2+- and DAG-driven signaling pathways have been less clearly defined.

In our recent work, we have identified Ca2+- and DAG-regulated guanine nucleotide exchange factor I (CalDAG-GEFI, RasGRP2) as a key molecule regulating Ca2+-dependent activation of integrins in platelets.16,17  CalDAG-GEFI contains binding sites for Ca2+ and DAG and a GEF domain catalyzing the activation of small GTPases of the Ras family, in particular Rap1 and Rap2.18  In platelets, Rap1B accounts for 90% of the total Rap protein,19  and its importance in αIIbβ3 activation has recently been demonstrated in Rap1B-deficient mice.20  Importantly, studies in cell types other than platelets demonstrated only very weak affinity of the C1 domain of CalDAG-GEFI for DAG in platelets,21,22  suggesting that it is predominantly regulated by binding of Ca2+ to its hand domains. Several of our recent studies with platelets isolated from CalDAG-GEFI–deficient mice support this assumption. First, platelet aggregation induced by Ca2+ ionophores, but not by the phorbol ester phorbol myristate acetate (DAG mimetic), was sensitive to CalDAG-GEFI expression.16  Second, CalDAG-GEFI was critical for the rapid, but reversible activation of Rap1 in thrombin-activated platelets, a process that depends on increased [Ca2+]i.17  And third, CalDAG-GEFI–dependent integrin activation occurred independent of signaling by PKC/P2Y12, suggesting that CalDAG-GEFI is part of the Ca2+-dependent pathway of platelet activation, which synergizes with signaling by PKC in integrin activation.14,15 

Rap1 has been extensively studied for its role in the regulation of cell adhesion.23  In cells other than platelets, Rap1 also regulates cellular differentiation and proliferation via pathways that involve the regulation of the mitogen-activated protein kinase (MAPK) cascade.24  Rap1 either interferes with Ras-mediated extracellular signal-regulated kinase (ERK) activation or it activates ERK independently of Ras in a cell context–dependent manner.25  In certain neurons, regulation of Rap1-dependent MAPK activation has been found to be directly downstream of CalDAG-GEFI.26,27  In platelets, ERK-MAPKs have been implicated in the phosphorylation of cytosolic phospholipase A2 (cPLA2),28  a key enzyme in the formation of arachidonic acid and thus TxA2.29-31  Interestingly, both Ca2+ and PKC signaling contribute to ERK1/2 activation.32 

In the present study, we investigated the mechanisms by which signaling by CalDAG-GEFI and PKC affects various aspects of platelet activation induced via stimulation of the collagen receptor, GPVI. Our studies demonstrate that CalDAG-GEFI is the main Ca2+ sensor that links increases in intracellular Ca2+ levels to the signaling pathways regulating integrin activation and ERK-MAPK signaling/TxA2 formation in stimulated platelets. CalDAG-GEFI mediates the first wave of platelet TxA2 formation, which provides critical feedback for the activation of PKC and granule release. P2Y12/Gi-dependent signaling induces a second wave of platelet activation, which is required for the formation of stable platelet aggregates.

Reagents and antibodies

Lovenox (enoxaparin sodium; Sanofi-Aventis), heparin-coated capillaries (VWR), bovine serum albumin (fraction V), prostacyclin, human fibrinogen (type I), and U0126 (mitogen-activated protein kinase kinase [MEK] inhibitor; all from Sigma-Aldrich), 2-methylthio-adenosine 5′-monophosphate triethylammonium salt hydrate (2-MesAMP, P2Y12 inhibitor; BioLog), complete protease inhibitor mixture lacking ethylenediaminetetraacetic acid (Roche Applied Science), Ro31-8220 (PKC inhibitor; EMD Chemicals), fibrillar collagen type I (Chronlog), calcein AM (Invitrogen), RalGDS-RBD coupled to agarose beads and polyvinylidene fluoride membranes (Millipore), U46619 (Cayman Chemical), acetylsalicylic acid (Bayer), and 3H-serotonin (Amersham Biosciences) were purchased. Convulxin (Cvx) was provided by K. Clemetson (Theodor Kocher Institute, University of Berne). Monoclonal blocking antibody to murine αIIbβ3, Leo.H4, and monoclonal antibody directed against the activated form of murine αIIbβ3, JON/A-PE, were purchased from Emfret Analytics. Anti-phospho(Thr202/Tyr204)-ERK, anti-ERK, and anti–phospho-(Ser) PKC substrate antibody were purchased from Cell Signaling Technology. Anti-Rap1 was purchased from Santa Cruz Biotechnology.

Mice

CalDAG-GEFI−/− mice16  on a C57B1/6 background were obtained from the lab of Ann Graybiel (Massachusetts Institute of Technology) and were bred in the mouse facility of Thomas Jefferson University. Experimental procedures were approved by the Animal Care and Use Committee of Thomas Jefferson University.

Platelet preparation

Blood was drawn from the retroorbital plexus into heparinized tubes. Platelet-rich plasma was obtained by centrifugation at 100g for 5 minutes. Platelet-rich plasma was centrifuged at 700g in the presence of prostacyclin (2 μg/mL) for 5 minutes at room temperature. After 2 washing steps, pelleted platelets were resuspended at the concentration of 4 × 108 platelets/mL in modified Tyrode buffer (137 mM NaCl, 0.3 mM Na2HPO4, 2 mM KCl, 12 mM NaHCO3, 5 mM N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid, 5 mM glucose, pH 7.3) containing 0.35% bovine serum albumin and 1 mM CaCl2.

Flow cytometry

Washed platelets were diluted in Tyrode buffer containing 1 mM CaCl2, activated with Cvx and/or U46619 in the presence of JON/A-PE33  for 10 minutes, and studied immediately by flow cytometry. MesAMP was added before platelet stimulation to block P2Y12 receptor signaling.

Aggregometry

Light transmission was measured in washed platelets activated in the presence of 50 μg/mL fibrinogen under stirring conditions at 37°C. Inhibitors and agonists were added at the indicated concentrations, and light transmission was recorded more than 10 minutes on a Chrono-log 4-channel optical aggregation system (Chrono-log).

Thromboxane generation assay

Platelets were stimulated in standard aggregometry. At different time points, 50 μL of sample was withdrawn and 5 mM EDTA (ethylenediaminetetraacetic acid)/1 mM aspirin was added. The cells were removed by centrifugation, and thromboxane B2 (TxB2) levels were determined in the supernatant by enzyme immunoassay (Assay Designs). TxB2 is the stable product of the nonenzymatic hydration of TxA2, which itself has a half-life of only 37 seconds under physiologic conditions.

Serotonin release

Washed platelets were incubated for 30 minutes at 37°C with 3H-serotonin (2 μCi [0.074 MBg]/mL). After one washing step, platelets were resuspended (4 × 108 platelets/mL) in modified Tyrode buffer containing 1 μM imipramine and 1 mM CaCl2. Platelets were stimulated in standard aggregometry. At the indicated time points, 50 μL of sample was withdrawn, and the reaction was stopped with an equal volume of 0.1 M EDTA/2% formaldehyde. The samples were then centrifuged for 5 minutes at 10 000g, and the supernatants were used for scintillation counting of 3H-serotonin. Total or 100% 3H-serotonin secretion was defined as the 3H-serotonin in samples lysed with 0.5% Triton X-100.

Rap1 and ERK activation

Platelets were stimulated in standard aggregometry for various times. Reactions were stopped with ice-cold 2× lysis buffer (100 mM Tris/HCl, pH 7.4, 400 mM NaCl, 5 mM MgCl2, 2% Nonidet P-40, 20% glycerol, and complete protease inhibitor mixture lacking ethylenediaminetetraacetic acid). Cell lysis was completed on ice for 15 minutes. A total of 200 μL of sample was immediately solubilized in sample buffer (75 mM Tris/HCl, pH 6.8, 10% sodium dodecyl sulfate, 5% 2-mercaptoethanol, 0.004% bromphenol blue) for the detection of phosphorylated ERK2 and total ERK1/2 levels. A total of 200 μL of lysate was used to detect activated Rap1 according to the protocol previously described.16  Briefly, Rap1-adenosine triphosphate (GTP) was precipitated from lysates using RalGDS-RBD beads (Millipore).

Western blotting

Total protein lysates and precipitated proteins were separated on 4% to 12% or 4% to 20% sodium dodecyl sulfate–polyacrylamide gel electrophoresis gradient gels and transferred to polyvinylidene fluoride membranes. Phosphorylated and nonphosphorylated ERK1/2 and Rap1 were detected with rabbit polyclonal antibodies. PKC-dependent protein phosphorylation was evaluated using a rabbit anti–phospho-(Ser) PKC substrate antibody. After incubation with anti–rabbit antibodies, conjugated to horseradish peroxidase (Vector Laboratories), immunoreactivity was detected by Western Lightning enhanced chemiluminescence (G-Biosciences).

Flow chamber assay

Whole blood anticoagulated with heparin (30 U/mL low molecular weight heparin; Lovenox) was perfused at a shear rate of 400 seconds−1 over bovine type I collagen–coated glass slides (PureCol, 300 μg/mL; Inamed Biomaterials) in a parallel-plate flow chamber. Platelet adhesion was visualized with a 20×/0.45 PlanFluor objective on a Nikon Ti-U inverted microscope (Nikon Instruments Inc) equipped with a Retiga EXL monochrome camera (QImaging). Images were analyzed using Nikon NIS Elements AR 3.0 software and processed with Photoshop CS3. After 5 minutes of perfusion, the number of firmly adherent platelets was determined in 3 separate areas (0.02 mm2). The experiment was repeated 3 times for each genotype.

Statistics

Results are reported as mean plus or minus SEM. Statistical significance was assessed by unpaired 2-tailed Student t test. A P value less than .05 was considered significant.

The generation of TxA2 and ADP is crucial for platelet aggregation in response to stimulation of the main collagen receptor, GPVI. To test whether CalDAG-GEFI is critical for GPVI-induced formation of second wave mediators, we first studied TxA2 generation in CalDAG-GEFI−/− platelets activated with the nonphysiologic agonist. As shown in Figure 1A, TxA2 generation, as measured by the formation of its stable analog TxB2, was completely abolished in knockout platelets stimulated with low dose Cvx (100 ng/mL). In response to high dose (500 ng/mL) of the agonist, TxA2 production was delayed and reached approximately 3-fold lower concentrations in CalDAG-GEFI−/− platelets compared with WT (Figure 1B).

Figure 1

CalDAG-GEFI is central to Cvx-dependent TxA2 generation. Washed platelets in the presence of 1 mM Ca2+ and 50 μg/mL fibrinogen were stimulated under stirring conditions at 37°C (standard aggregometry) with (A,C) low (LD, 100 ng/mL) or (B,D) high (HD, 500 ng/mL) dose Cvx. Samples were withdrawn at the indicated time points to measure the levels of TxB2, the stable product of the nonenzymatic hydration of TxA2. (A-B) TxB2 levels in the supernatant of WT (black line, ●) or CalDAG-GEFI−/− (knockout [KO], gray line, ▴) platelets activated in the presence (open symbols) or absence (filled symbols) of 75 μM 2-MesAMP (P2Y12 inhibitor). Data shown are mean ± SEM (n = 4). At a high dose of Cvx (B), significantly higher levels of TxB2 were detected at all time points in the supernatant of activated WT platelets compared with WT/MesAMP, KO, or KO/MesAMP platelets. (C-D) Aggregation traces representative of 3 independent experiments. (E) WT (■) or CalDAG-GEFI–deficient (▩) platelets were stimulated with LD (100 ng/mL) or HD (750 ng/mL) Cvx in the presence or absence of 2-MesAMP. Binding of JON/A-PE was measured to determine the level of αIIbβ3 activation by flow cytometry. n = 6. (F) TxB2 release from WT and CalDAG-GEFI−/− (KO) platelets stimulated for 10 minutes with HD Cvx in the presence (□) or absence (■) of 40 μg/mL αIIbβ3 blocking antibody Leo.H4. Data shown are mean ± SEM (n = 3), *P < .05, **P < .01, ***P < .001.

Figure 1

CalDAG-GEFI is central to Cvx-dependent TxA2 generation. Washed platelets in the presence of 1 mM Ca2+ and 50 μg/mL fibrinogen were stimulated under stirring conditions at 37°C (standard aggregometry) with (A,C) low (LD, 100 ng/mL) or (B,D) high (HD, 500 ng/mL) dose Cvx. Samples were withdrawn at the indicated time points to measure the levels of TxB2, the stable product of the nonenzymatic hydration of TxA2. (A-B) TxB2 levels in the supernatant of WT (black line, ●) or CalDAG-GEFI−/− (knockout [KO], gray line, ▴) platelets activated in the presence (open symbols) or absence (filled symbols) of 75 μM 2-MesAMP (P2Y12 inhibitor). Data shown are mean ± SEM (n = 4). At a high dose of Cvx (B), significantly higher levels of TxB2 were detected at all time points in the supernatant of activated WT platelets compared with WT/MesAMP, KO, or KO/MesAMP platelets. (C-D) Aggregation traces representative of 3 independent experiments. (E) WT (■) or CalDAG-GEFI–deficient (▩) platelets were stimulated with LD (100 ng/mL) or HD (750 ng/mL) Cvx in the presence or absence of 2-MesAMP. Binding of JON/A-PE was measured to determine the level of αIIbβ3 activation by flow cytometry. n = 6. (F) TxB2 release from WT and CalDAG-GEFI−/− (KO) platelets stimulated for 10 minutes with HD Cvx in the presence (□) or absence (■) of 40 μg/mL αIIbβ3 blocking antibody Leo.H4. Data shown are mean ± SEM (n = 3), *P < .05, **P < .01, ***P < .001.

Close modal

Previous studies demonstrated that ADP/P2Y12 signaling is a major component of thrombin-induced TxA2 generation.31  Thus, we investigated the role of the ADP/P2Y12 signaling pathway downstream of the collagen receptor GPVI. The P2Y12 inhibitor, 2-MesAMP, impaired TxA2 release from Cvx-activated WT platelets to a similar extent as the absence of CalDAG-GEFI. Moreover, 2-MesAMP completely abolished the residual TxA2 release observed in Cvx-stimulated CalDAG-GEFI−/− platelets (Figure 1B), demonstrating a synergistic role of P2Y12–dependent and CalDAG-GEFI–dependent signaling in GPVI-dependent TxA2 generation.

The time course of TxA2 generation showed a striking correlation with integrin activation as monitored by standard aggregometry (Figure 1C-D). Direct assessment of αIIbβ3 activation in platelets stimulated with low dose Cvx, using an antibody against the activated form of the receptor (JON/A-PE33 ), demonstrated completely inhibited activation of αIIbβ3 in CalDAG-GEFI−/− platelets (Figure 1E). At high dose of Cvx, CalDAG-GEFI−/− platelets showed a small, but significant increase in JON/A-PE binding, which was abolished in the presence of 2-MesAMP. To exclude the possibility that the defect in TxA2 generation was secondary to the defect in integrin activation observed in knockout platelets, we determined TxA2 release from platelets activated in the presence or absence of a blocking antibody to αIIbβ3. Integrin inhibition marginally reduced TxA2 production in both WT and CalDAG-GEFI−/− platelets. However, there was a significant difference in TxA2 release between CalDAG-GEFI–deficient and αIIbβ3-inhibited WT platelets (P < .05; Figure 1F), demonstrating that CalDAG-GEFI contributes to TxA2 generation independent of its role in integrin inside-out signaling.

To further characterize the signaling pathways that lead to CalDAG-GEFI–dependent and CalDAG-GEFI–independent TxA2 generation, we compared the kinetics of Rap1 activation with that of ERK-MAPK phosphorylation. Both ERK phosphorylation and Rap1 activation were completely abolished in CalDAG-GEFI−/− platelets activated with 100 ng/mL Cvx (Figure 2A). When activated with 500 ng/mL Cvx, activation of both Rap1 and ERK was delayed (Figure 2B), and the residual activation observed in knockout platelets was abolished upon pretreatment of the cells with 2-MesAMP (Figure 2C).

Figure 2

CalDAG-GEFI and P2Y12 synergize in Cvx-dependent activation of Rap1 and ERK-MAPK. Washed WT or CalDAG-GEFI−/− (knockout [KO]) platelets were stimulated in standard aggregometry with (A) 100 ng/mL (low dose [LD]) or (B-C) 500 ng/mL (high dose [HD]) Cvx. Platelets were activated in the presence (C) of 75 μM 2-MesAMP to inhibit signaling by P2Y12. Rap1-GTP and phospho-ERK were determined in samples withdrawn at the indicated time points. Total ERK1/2 levels (bottom panel) were determined as a loading control. Results are representative of 3 individual experiments.

Figure 2

CalDAG-GEFI and P2Y12 synergize in Cvx-dependent activation of Rap1 and ERK-MAPK. Washed WT or CalDAG-GEFI−/− (knockout [KO]) platelets were stimulated in standard aggregometry with (A) 100 ng/mL (low dose [LD]) or (B-C) 500 ng/mL (high dose [HD]) Cvx. Platelets were activated in the presence (C) of 75 μM 2-MesAMP to inhibit signaling by P2Y12. Rap1-GTP and phospho-ERK were determined in samples withdrawn at the indicated time points. Total ERK1/2 levels (bottom panel) were determined as a loading control. Results are representative of 3 individual experiments.

Close modal

In addition to Cvx, we studied platelets stimulated with fibrillar type I collagen (Figure 3). Collagen activation led to approximately 40% more TxA2 generation in WT platelets compared with Cvx. TxA2 release in collagen-activated CalDAG-GEFI–deficient platelets was markedly impaired and delayed (Figure 3A). The kinetics of TxA2 release correlated well with those of Rap1 and ERK activation (Figure 3C), suggesting that CalDAG-GEFI is crucial for the first wave of TxA2 release. Pretreatment of knockout platelets with 2-MesAMP markedly reduced, but did not abolish collagen-induced TxA2 release (Figure 3A), although activation of both ERK and Rap1 was completely inhibited under these experimental conditions (Figure 3D).

Figure 3

CalDAG-GEFI is critical for collagen-dependent TxA2 generation. (A) Time course of TxB2 release from WT (black line, circle) or CalDAG-GEFI−/− (knockout [KO], gray line, triangle) platelets. Cells were stimulated with 25 μg/mL fibrillar type I collagen under stirring conditions in the presence (open symbols) or absence (filled symbols) of 75 μM 2-MesAMP. Data shown are mean ± SEM (n = 4). Significantly higher levels of TxB2 were detected at indicated time points in the supernatant of activated WT platelets compared with KO or KO/MesAMP platelets. (B) Aggregation traces representative of 3 independent experiments. (C-D) Time course of ERK (top panel) and Rap1 (middle panel) activation in platelets activated with collagen in the presence (D) or absence (C) of 75 μM 2-MesAMP. Representative of 3 independent experiments. (E) TxB2 levels in the supernatant of WT platelets stimulated for 10 minutes with 500 ng/mL Cvx (high dose [HD] Cvx), or 25 μg/mL fibrillar collagen (HD collagen). Cells were activated in the presence (□) or absence (■) of 50 μM U0126 (MEK inhibitor). U0126 totally abolished ERK phosphorylation in response to Cvx or collagen (data not shown). Data are shown as mean ± SEM. The amount of TxB2 released from collagen-activated platelets was defined as 100%. n = 3. *P < .05, **P < .01, ***P < .001.

Figure 3

CalDAG-GEFI is critical for collagen-dependent TxA2 generation. (A) Time course of TxB2 release from WT (black line, circle) or CalDAG-GEFI−/− (knockout [KO], gray line, triangle) platelets. Cells were stimulated with 25 μg/mL fibrillar type I collagen under stirring conditions in the presence (open symbols) or absence (filled symbols) of 75 μM 2-MesAMP. Data shown are mean ± SEM (n = 4). Significantly higher levels of TxB2 were detected at indicated time points in the supernatant of activated WT platelets compared with KO or KO/MesAMP platelets. (B) Aggregation traces representative of 3 independent experiments. (C-D) Time course of ERK (top panel) and Rap1 (middle panel) activation in platelets activated with collagen in the presence (D) or absence (C) of 75 μM 2-MesAMP. Representative of 3 independent experiments. (E) TxB2 levels in the supernatant of WT platelets stimulated for 10 minutes with 500 ng/mL Cvx (high dose [HD] Cvx), or 25 μg/mL fibrillar collagen (HD collagen). Cells were activated in the presence (□) or absence (■) of 50 μM U0126 (MEK inhibitor). U0126 totally abolished ERK phosphorylation in response to Cvx or collagen (data not shown). Data are shown as mean ± SEM. The amount of TxB2 released from collagen-activated platelets was defined as 100%. n = 3. *P < .05, **P < .01, ***P < .001.

Close modal

To confirm the role of ERK signaling in Rap1-dependent TxA2 generation, we compared our results with 2-MesAMP–pretreated CalDAG-GEFI−/− platelets (Rap1 activation completely blocked) with those obtained with WT platelets pretreated with the MEK inhibitor, U0126.34  Pretreatment with U0126 completely inhibited ERK phosphorylation (not shown) and TxA2 release in Cvx-activated WT platelets (Figure 3E). In collagen-activated platelets, U0126 abolished ERK phosphorylation (not shown), whereas it reduced TxA2 release by approximately 60%. Thus, the GPVI/Rap1/ERK-dependent signaling pathway accounts for approximately 60% of the TxA2 production in collagen-activated platelets.

Based on these results, we speculated that the defect in TxA2 generation explains why CalDAG-GEFI−/− platelets fail to aggregate in response to low concentrations of Cvx, and that addition of exogenous TxA2 could restore aggregation of Cvx-stimulated knockout platelets. As expected, aggregation was observed in CalDAG-GEFI−/− platelets stimulated with threshold concentrations of the thromboxane mimetic, U46619, and low dose Cvx, but not with either agonist alone (Figure 4A). Aggregation was reversed by pretreatment with a broad range PKC inhibitor, Ro31-8220, or the P2Y12 inhibitor 2-MesAMP. Consequently, binding of JON/A-PE to mutant platelets costimulated with Cvx and U46619 was significantly higher than in mutant platelets activated with either agonist alone, or platelets activated with Cvx and U46619 in the presence of 2-MesAMP (Figure 4B). Thus, in CalDAG-GEFI−/− platelets activated with a low dose of a GPVI agonist, TxA2 is required to trigger integrin activation through the CalDAG-GEFI–independent, but PKC/P2Y12–dependent pathway.

Figure 4

Exogenous TxA2 restores aggregation, PKC activation, and dense granule secretion in Cvx-stimulated CalDAG-GEFI−/− platelets. (A) Left: Aggregation of WT platelets stimulated with low dose Cvx (100 ng/mL). Right: Aggregation traces for CalDAG-GEFI−/− platelets activated with 100 ng/mL Cvx, 500 nM U46619, or the combination of both agonists. Platelet aggregation was studied in the presence of 75 μM 2-MesAMP or 5 μg/mL Ro31-8220 to block signaling by P2Y12 and PKC, respectively. Results are representative of 5 individual experiments. (B) CalDAG-GEFI–deficient platelets were stimulated with low dose (LD) Cvx (100 ng/mL) and/or U46619 (3 μM) in the presence or absence of 2-MesAMP. Binding of JON/A-PE was measured to determine the level of αIIbβ3 activation by flow cytometry. n = 6. **P < .001, *P < .05. (C) Detection of phosphorylated PKC substrates in platelet lysates (Western blotting). WT (left lanes) and CalDAG-GEFI−/− (knockout [KO], central lanes) platelets were stimulated for 10 minutes with 100 ng/mL Cvx, 500 nM U46619, or the combination of both agonists. Phosphorylation of PKC substrates, including PLEK (←), in WT platelets pretreated with Ro31-8220 (10 μg/mL) or activated with phorbol myristate acetate (100 nM) were determined as controls (right panel). Results are representative of 3 independent experiments. (D) 3H-serotonin release in WT (black line, ●) or CalDAG-GEFI−/− (gray line, gray triangle) platelets activated with 100 ng/mL Cvx. After 3 minutes, 500 nM U46619 was added to CalDAG-GEFI−/− platelets. Experiments were performed in the presence (open symbols) or absence (filled symbols) of the PKC inhibitor Ro31-8220 (n = 3).

Figure 4

Exogenous TxA2 restores aggregation, PKC activation, and dense granule secretion in Cvx-stimulated CalDAG-GEFI−/− platelets. (A) Left: Aggregation of WT platelets stimulated with low dose Cvx (100 ng/mL). Right: Aggregation traces for CalDAG-GEFI−/− platelets activated with 100 ng/mL Cvx, 500 nM U46619, or the combination of both agonists. Platelet aggregation was studied in the presence of 75 μM 2-MesAMP or 5 μg/mL Ro31-8220 to block signaling by P2Y12 and PKC, respectively. Results are representative of 5 individual experiments. (B) CalDAG-GEFI–deficient platelets were stimulated with low dose (LD) Cvx (100 ng/mL) and/or U46619 (3 μM) in the presence or absence of 2-MesAMP. Binding of JON/A-PE was measured to determine the level of αIIbβ3 activation by flow cytometry. n = 6. **P < .001, *P < .05. (C) Detection of phosphorylated PKC substrates in platelet lysates (Western blotting). WT (left lanes) and CalDAG-GEFI−/− (knockout [KO], central lanes) platelets were stimulated for 10 minutes with 100 ng/mL Cvx, 500 nM U46619, or the combination of both agonists. Phosphorylation of PKC substrates, including PLEK (←), in WT platelets pretreated with Ro31-8220 (10 μg/mL) or activated with phorbol myristate acetate (100 nM) were determined as controls (right panel). Results are representative of 3 independent experiments. (D) 3H-serotonin release in WT (black line, ●) or CalDAG-GEFI−/− (gray line, gray triangle) platelets activated with 100 ng/mL Cvx. After 3 minutes, 500 nM U46619 was added to CalDAG-GEFI−/− platelets. Experiments were performed in the presence (open symbols) or absence (filled symbols) of the PKC inhibitor Ro31-8220 (n = 3).

Close modal

To confirm this hypothesis, we evaluated the phosphorylation state of downstream targets of PKC, such as pleckstrin-1 (PLEK),35  in stimulated platelets. In WT platelets, PLEK phosphorylation was observed in response to threshold doses of Cvx or U46619. In contrast, PLEK phosphorylation was markedly reduced in CalDAG-GEFI–deficient platelets activated with low dose Cvx or low dose U46619. However, PLEK phosphorylation was restored in CalDAG-GEFI−/− platelets activated with both Cvx and U46619 (Figure 4C). To test whether impaired integrin activation at low Cvx concentrations could be the result of defective release and cosignaling by ADP, we next studied dense granule release in CalDAG-GEFI–deficient platelets. Release of 3H-labeled serotonin from platelets activated under stirring conditions was measured (Figure 4D). Low dose Cvx stimulation induced the rapid release of serotonin from WT, but not CalDAG-GEFI−/−, platelets. However, a threshold dose of U46619 could partially restore Cvx-induced secretion. Secretion under these conditions was dependent on PKC activation, as it was blocked by the broad range PKC inhibitor, Ro31-8220.

Based on these findings, we speculated that TxA2 released from WT platelets might be able to induce the aggregation of CalDAG-GEFI−/− platelets activated with low dose collagen. As shown in Figure 5A, addition of 10% WT platelets partially restored aggregation of CalDAG-GEFI−/− platelets (62% ± 23% of WT). In contrast, WT platelets pretreated with aspirin were unable to restore aggregation of knockout platelets (0% of WT). Furthermore, 10% of WT platelets failed to restore aggregation of CalDAG-GEFI−/− platelets pretreated with a αIIbβ3 blocking antibody (5% ± 1% of WT). Thus, TxA2 released from the WT platelets provided a stimulatory signal required for αIIbβ3 activation in CalDAG-GEFI−/− platelets activated with low dose collagen. To confirm the generation of aggregates consisting of both WT and CalDAG-GEFI−/− platelets, we repeated these studies with calcein green-labeled WT and calcein red-labeled CalDAG-GEFI−/− platelets (Figure 5B). Fixed aggregates were visualized by fluorescence microscopy. As expected, green-labeled WT platelets recruited a large number of red-labeled CalDAG-GEFI−/− platelets into the aggregates. Pretreatment with an αIIbβ3 blocking antibody, however, almost completely blocked the incorporation of knockout platelets.

Figure 5

TxA2 released from WT platelets restores aggregation of CalDAG-GEFI−/− platelets. (A) Aggregation traces of platelets (108 cells) activated with 10 μg/mL collagen. The aggregation response of WT (black line), CalDAG-GEFI−/− (knockout [KO], light gray line), or CalDAG-GEFI−/− platelets containing 10% WT platelets (dark gray line) was studied. WT platelets were pretreated with 1 mM aspirin (acetylsalicylic acid) before addition to CalDAG-GEFI−/− platelets to inhibit TxA2 release. CalDAG-GEFI−/− platelets were pretreated with 40 μg/mL blocking antibody to αIIbβ3 to demonstrate that the aggregation was integrin-dependent. Traces are representative of 3 individual experiments. (B) Representative images showing aggregates of calcein green–labeled WT and/or calcein red–labeled KO (CalDAG-GEF−/−) platelets fixed with 3.7% formaldehyde 10 minutes after stimulation and visualized with fluorescence microscopy. Scale bar = 25 μm.

Figure 5

TxA2 released from WT platelets restores aggregation of CalDAG-GEFI−/− platelets. (A) Aggregation traces of platelets (108 cells) activated with 10 μg/mL collagen. The aggregation response of WT (black line), CalDAG-GEFI−/− (knockout [KO], light gray line), or CalDAG-GEFI−/− platelets containing 10% WT platelets (dark gray line) was studied. WT platelets were pretreated with 1 mM aspirin (acetylsalicylic acid) before addition to CalDAG-GEFI−/− platelets to inhibit TxA2 release. CalDAG-GEFI−/− platelets were pretreated with 40 μg/mL blocking antibody to αIIbβ3 to demonstrate that the aggregation was integrin-dependent. Traces are representative of 3 individual experiments. (B) Representative images showing aggregates of calcein green–labeled WT and/or calcein red–labeled KO (CalDAG-GEF−/−) platelets fixed with 3.7% formaldehyde 10 minutes after stimulation and visualized with fluorescence microscopy. Scale bar = 25 μm.

Close modal

To validate our findings under conditions of physiologic flow, we tested the effect of exogenous thromboxane on the adhesion of CalDAG-GEFI−/− platelets to collagen in flow chamber studies (Figure 6A). Whereas WT platelets formed thrombi on the collagen surface, only few CalDAG-GEFI−/− platelets were able to firmly adhere to collagen under these conditions. Addition of U46619 led to significantly more firm adhesion of CalDAG-GEFI−/− platelets (Figure 6B). The effect of U46619 was reversed by pretreating CalDAG-GEFI−/− platelets with 2-MesAMP, suggesting that exogenous thromboxane promotes adhesion of knockout platelets via an ADP/P2Y12-dependent mechanism.

Figure 6

Exogenous TxA2 induces firm adhesion of CalDAG-GEFI−/− platelets to collagen. Whole blood from WT or CalDAG-GEFI−/− mice was perfused over a collagen surface at venous shear rates (400 seconds−1). (A) Representative images showing platelets firmly adherent to the collagen surface. U46619 (2 μM) was added to CalDAG-GEFI−/− (knockout [KO]) whole blood immediately before the perfusion. A total of 100 μM 2-MesAMP was added 10 minutes before the perfusion. Scale bar = 10 μm. (B) Quantification of adherent platelets. The data are presented as the mean ± SEM of the absolute number of platelets per 0.02 mm2 surface area. n = 9; 3 different blood preparations. **P < .01.

Figure 6

Exogenous TxA2 induces firm adhesion of CalDAG-GEFI−/− platelets to collagen. Whole blood from WT or CalDAG-GEFI−/− mice was perfused over a collagen surface at venous shear rates (400 seconds−1). (A) Representative images showing platelets firmly adherent to the collagen surface. U46619 (2 μM) was added to CalDAG-GEFI−/− (knockout [KO]) whole blood immediately before the perfusion. A total of 100 μM 2-MesAMP was added 10 minutes before the perfusion. Scale bar = 10 μm. (B) Quantification of adherent platelets. The data are presented as the mean ± SEM of the absolute number of platelets per 0.02 mm2 surface area. n = 9; 3 different blood preparations. **P < .01.

Close modal

Our studies establish a revised model for platelet activation by collagen (Figure 7). The key elements of this new model are as follows: (1) the central role of one molecule, CalDAG-GEFI, in Ca2+-dependent platelet activation; (2) the preferential activation of CalDAG-GEFI over PKC downstream of PLC activation; (3) the importance of Rap1, activated by CalDAG-GEFI and P2Y12 signaling, in integrin activation and TxA2 generation; (4) the critical role of CalDAG-GEFI in TxA2 release and subsequent PKC activation, degranulation, and activation of P2Y12/Gi signaling; and (5) the kinetic differences between CalDAG-GEFI– and P2Y12–mediated Rap1 activation and the respective downstream signaling events.

Figure 7

A revised model of platelet activation in response to low dose collagen. (1) A rise in [Ca2+]i stimulates the CalDAG-GEFI–dependent first wave of Rap1 and integrin activation. Activated Rap1 also triggers ERK-dependent generation of TxA2, which in turn provides critical feedback for the activation of PKC. (2) In a next step, PKC triggers the release of granule contents, including ADP. Released ADP signals through P2Y12/Gi to stimulate the second wave of Rap1/integrin activation and TxA2 generation, which is required for the formation of stable platelet-platelet contacts. Higher concentrations of GPVI agonists lead to the concomitant activation of CalDAG-GEFI and PKC, facilitating integrin activation independent of feedback by endogenous TxA2 (dashed line). AA indicates arachidonic acid; Gi, heterotrimeric G protein; P, residue phosphorylation; PL, phospholipids; and TPα/β, thromboxane receptors.

Figure 7

A revised model of platelet activation in response to low dose collagen. (1) A rise in [Ca2+]i stimulates the CalDAG-GEFI–dependent first wave of Rap1 and integrin activation. Activated Rap1 also triggers ERK-dependent generation of TxA2, which in turn provides critical feedback for the activation of PKC. (2) In a next step, PKC triggers the release of granule contents, including ADP. Released ADP signals through P2Y12/Gi to stimulate the second wave of Rap1/integrin activation and TxA2 generation, which is required for the formation of stable platelet-platelet contacts. Higher concentrations of GPVI agonists lead to the concomitant activation of CalDAG-GEFI and PKC, facilitating integrin activation independent of feedback by endogenous TxA2 (dashed line). AA indicates arachidonic acid; Gi, heterotrimeric G protein; P, residue phosphorylation; PL, phospholipids; and TPα/β, thromboxane receptors.

Close modal

In platelets activated with threshold concentrations of GPVI agonists, CalDAG-GEFI serves as a highly sensitive response element to Ca2+ that allows for the rapid activation of Rap1. CalDAG-GEFI–mediated Rap1 activation triggers a first wave of integrin activation and ERK (MAPK) signaling, followed by TxA2 release. TxA2 provides crucial feedback for the activation of PKC and granule/ADP release. ADP in turn triggers the second, P2Y12-dependent wave of Rap1-mediated signaling events, leading to the sustained activation of integrins and further release of TxA2. Higher concentrations of GPVI agonists lead to the concomitant activation of CalDAG-GEFI and PKC, facilitating integrin activation independent of feedback by endogenous TxA2. It is important to note that the Gαq-coupled receptor for ADP, P2Y1,36  also contributes to PLC signaling, thus supporting both CalDAG-GEFI– and PKC/P2Y12–dependent platelet activation (data not shown).

Our studies identify CalDAG-GEFI as a long sought-after link between increased [Ca2+]i and the signaling mechanisms regulating integrin activation and TxA2 release. To ensure an independent role of CalDAG-GEFI in both processes, we blocked integrin outside-in signaling or TxA2 generation in platelets. The αIIbβ3 blocking antibody, Leo.H4, only marginally reduced TxA2 production in WT or knockout platelets (Figure 1F) and did not have any effect on ERK phosphorylation, demonstrating that CalDAG-GEFI–dependent TxA2 production is independent of CalDAG-GEFI–mediated integrin activation. In contrast, the defect in integrin activation in CalDAG-GEFI−/− platelets was in part secondary to impaired TxA2 generation observed in these cells. At high agonist concentrations, aggregation was mediated by CalDAG-GEFI and P2Y12 signaling in a TxA2-independent manner. Aggregation of knockout platelets activated with low dose Cvx or low dose collagen, however, was restored by addition of exogenous TxA2 (Figures 4A and 5). TxA2 added important feedback for the activation of PKC and thus granule/ADP release (Figure 4C-D).

The small GTPase Rap1 is the major substrate of CalDAG-GEFI in platelets. We and others have demonstrated that CalDAG-GEFI, P2Y12, and Rap1 are critical to integrin activation in platelets5,17,20,37  and megakaryocytes.38,39  In other cell types, Rap1 also regulates cellular differentiation and proliferation via pathways that involve the regulation of the MAPK cascade.24,25  In platelets, ERK-MAPK play a critical role in the activation of cPLA2,30,31  a key enzyme for the generation of arachidonic acid and thus TxA2.29  Our results provide the first evidence of a Rap1/ERK signaling pathway in platelets and of its involvement in TxA2 generation. We found that GPVI-dependent TxA2 generation relies entirely on the Rap1/ERK pathway, as inhibition of Rap1 activation (CalDAG-GEFI−/− platelets + 2-MesAMP) or ERK signaling (WT platelets + U0126) abolished Cvx-dependent TxA2 generation (Figure 3E). CalDAG-GEFI deficiency or inhibition of P2Y12 alone reduced TxA2 release by approximately 70%. However, a marked difference in the kinetics of CalDAG-GEFI– and P2Y12–mediated ERK activation was observed. CalDAG-GEFI facilitated the rapid, but reversible activation of ERK, whereas P2Y12 signaling led to a delayed, but sustained generation of phospho-ERK. The latter findings confirm recent studies that identified a critical role of P2Y12 signaling for ERK activation and TxA2 generation in thrombin-activated platelets.31 

Recent studies by Ginsberg and colleagues identified Rap1-interacting adapter molecule as the potential downstream effector of Rap1 that controls integrin activation in platelets. Their results further suggest that Rap1-interacting adapter molecule targets talin to the cytoplasmic domain of the integrin, a critical event in the activation of these adhesion receptors.40,41  Little is known about the downstream target of Rap1 that mediates the activation of ERK. In fibroblasts, Rap1 was shown to antagonize MAPK signaling by sequestering the serine/threonin kinase, C-Raf.42,43  In contrast, studies in neurons demonstrated that recruitment of B-Raf to Rap1-GTP results in the sequential activation of MEK and ERK.44  Thus, the activating or inhibitory effect of Rap1 on ERK signaling depends on the relative abundance of B-Raf and C-Raf found in a particular cell type.25,26,45  B-Raf is expressed in megakaryocytes and platelets, and the B-Raf/Rap1 pathway was shown to stimulate the ERK cascade in a model of megakaryocyte differentiation.46  C-Raf expression in platelets has also been demonstrated.47  Our studies suggest that Rap1/B-Raf/ERK signaling may be the predominant pathway in platelets. To our knowledge, there has been only one study on the role of Raf kinases in platelet activation, which suggested that ERK activation in response to thrombin relies on PKC, but not on Raf kinases.32  This conclusion, however, was based on an in vitro kinase activity assay, but not on the functional blockade of the kinases. Furthermore, these studies do not exclude the possibility of a B-Raf–dependent activation in platelets stimulated by agonists other than thrombin. Further studies are necessary to understand the role of Raf kinases in Rap1-dependent MAPK activation in platelets.

In addition to the GPVI-specific agonist Cvx, we have also stimulated platelets with fibrillar type I collagen, a physiologic agonist that binds to both GPVI and integrin α2β1. In contrast to Cvx-activated platelets, collagen-dependent TxA2 release was only approximately 60% reduced in 2-MesAMP–treated CalDAG-GEFI−/− platelets or MEK/ERK inhibitor-treated WT platelets. Correlating with these results, we observed that WT platelets stimulated with fibrillar collagen produced approximately 40% more TxA2 compared with cells activated with high dose Cvx. The most likely explanation for the Rap1- and ERK-independent formation of TxA2 is outside-in signaling provided by α2β1. Several in vitro and in vivo studies suggested a supportive role of α2β1 in collagen-induced TxA2 formation and platelet activation.48,49  Notably, Heemskerk and colleagues50  demonstrated that platelets from α2 integrin-deficient mice and aspirin-treated WT platelets have similar defects in their adhesion to collagen under flow. Based on their results, they concluded that integrin α2β1 plays a role in stabilizing the thrombi by enhancing GPVI-dependent TxA2 release.50  In addition to ERK, the MAPK family members c-Jun N-terminal kinase 1 and p38 are present in platelets.51-53  Whereas p38 signaling has been linked to cPLA2 phosphorylation in platelets stimulated by various agonists,54,55  no such role has been described for c-Jun N-terminal kinase 1. In addition to MAPK signaling, Shattil and colleagues recently demonstrated that integrin outside-in signaling could directly affect the enzymatic activity of a pool of cPLA2α bound to the integrin.56  Independent of the mechanisms regulating this second pathway for collagen-induced TxA2 formation, our studies demonstrate that GPVI-dependent TxA2 release requires signaling by CalDAG-GEFI and P2Y12, which synergize at the level of Rap1 and ERK activation.

A key finding of this study is the critical role of CalDAG-GEFI signaling in GPVI-dependent TxA2 release (Figure 1). Based on previous reports showing that collagen-induced aggregation strongly depends on feedback by endogenous TxA2,57,58  we speculated that the aggregation/adhesion defects of CalDAG-GEFI−/− platelets to collagen16  could be overcome by adding exogenous TxA2. Indeed, we were able to restore GPVI-dependent aggregation of CalDAG-GEFI−/− platelets by adding U46619 (Figure 4A) or TxA2 released from WT platelets (Figure 5). Furthermore, addition of U46619 significantly increased the ability of CalDAG-GEFI−/− platelets to firmly adhere to a collagen surface under physiologic flow conditions (Figure 6). Exogenous TxA2 supported CalDAG-GEFI–independent, but P2Y12–dependent platelet adhesion by providing critical feedback for GPVI-induced activation of PKC and thus granule/ADP release (Figure 4C-D). These findings may have important implications for the development of novel antiplatelet agents as well as for improving existing antiplatelet strategies. Our data identify CalDAG-GEFI as a powerful new target. Due to its central role in the early phase of platelet activation, inhibitors of CalDAG-GEFI will have a strong antithrombotic effect, comparable with those observed with αIIbβ3 inhibitors. On strong thrombogenic surfaces, however, inhibition of CalDAG-GEFI can be partially overcome by PKC/P2Y12 signaling, resulting in the firm adhesion of platelets, and thus reduced bleeding. Our studies also provide new insights on the mechanisms by which P2Y12 inhibitors like clopidogrel, currently one of the most successful antiplatelet strategies in the clinic,59  prevent thrombosis. Platelets treated with P2Y12 inhibitors rely on signaling by CalDAG-GEFI for Rap1 activation. CalDAG-GEFI–mediated Rap1 activation occurs rapidly, but it is reversible. Consequently, P2Y12 inhibitors markedly reduce GPVI-induced TxA2 release, and integrin activation is transient. Under flow conditions, transient Rap1 activation does not allow stable platelet aggregates to form.

Future studies should be directed toward a better understanding of how P2Y12 signaling leads to Rap1 activation. In addition to CalDAG-GEFI, the Rap1-GEFs CalDAG-GEFIII,60  PDZ-GEF1,60  and Epac1 (cyclic adenosine 5′-monophosphate–GEFI)61  have been identified in platelets and may thus be involved. Rap1 activation is also regulated by GTPase-activating proteins (GAPs). In platelets, Rap1GAP2 was recently identified.60  In other cell types, Gαi/o signaling was shown to promote ubiquitination and proteosomal degradation of Rap1GAP2, leading to enhanced Rap1 activity.62  However, the role of the above-mentioned RapGEFs or Rap1GAP2 in P2Y12/Gαi–dependent Rap1 activation in platelets has not been addressed. Another area of interest will be studies aimed at a better understanding of how Rap1 activation can be sustained in the absence of P2Y12 signaling, or how to speed up Rap1 activation in the absence of CalDAG-GEFI signaling. Success in these studies could lead to a marked improvement of antiplatelet therapy, as it would provide strategies for the development of an antidote to P2Y12 or CalDAG-GEFI inhibitors.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Michael Holinstat for helpful discussions.

This work was supported by Scientist Development Grant 0630044N from the American Heart Association (W.B.).

Contribution: L.S. designed the study, performed many of the experiments, and wrote the paper; R.C.R. maintained the mouse colony and helped with the experiments; and W.B. designed the study and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Wolfgang Bergmeier, Cardeza Foundation and Department of Medicine, Thomas Jefferson University, 803 Curtis, 1015 Walnut St, Philadelphia, PA 19007; e-mail: wolfgang.bergmeier@jefferson.edu.

1
Savage
 
B
Almus-Jacobs
 
F
Ruggeri
 
ZM
Specific synergy of multiple substrate-receptor interactions in platelet thrombus formation under flow.
Cell
1998
, vol. 
94
 (pg. 
657
-
666
)
2
Nieswandt
 
B
Brakebusch
 
C
Bergmeier
 
W
, et al. 
Glycoprotein VI but not α2β1 integrin is essential for platelet interaction with collagen.
EMBO J
2001
, vol. 
20
 (pg. 
2120
-
2130
)
3
FitzGerald
 
GA
Mechanisms of platelet activation: thromboxane A2 as an amplifying signal for other agonists.
Am J Cardiol
1991
, vol. 
68
 (pg. 
11B
-
15B
)
4
Cosemans
 
JM
Munnix
 
IC
Wetzker
 
R
Heller
 
R
Jackson
 
SP
Heemskerk
 
JW
Continuous signaling via PI3K isoforms β and γ is required for platelet ADP receptor function in dynamic thrombus stabilization.
Blood
2006
, vol. 
108
 (pg. 
3045
-
3052
)
5
Kamae
 
T
Shiraga
 
M
Kashiwagi
 
H
, et al. 
Critical role of ADP interaction with P2Y12 receptor in the maintenance of αIIbβ3 activation: association with Rap1B activation.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
1379
-
1387
)
6
Harper
 
MT
Poole
 
AW
Isoform-specific functions of protein kinase C: the platelet paradigm.
Biochem Soc Trans
2007
, vol. 
35
 (pg. 
1005
-
1008
)
7
Konopatskaya
 
O
Gilio
 
K
Harper
 
MT
, et al. 
PKCα regulates platelet granule secretion and thrombus formation in mice.
J Clin Invest
2009
, vol. 
119
 (pg. 
399
-
407
)
8
Chari
 
R
Getz
 
T
Nagy
 
B
, et al. 
Protein kinase Cδ differentially regulates platelet functional responses.
Arterioscler Thromb Vasc Biol
2009
, vol. 
29
 (pg. 
699
-
705
)
9
Nagy
 
B
Bhavaraju
 
K
Getz
 
T
Bynagari
 
YS
Kim
 
S
Kunapuli
 
SP
Impaired activation of platelets lacking protein kinase Cθ isoform.
Blood
2009
, vol. 
113
 (pg. 
2557
-
2567
)
10
Bynagari
 
YS
Nagy
 
B
Tuluc
 
F
, et al. 
Mechanism of activation and functional role of protein kinase Cη in human platelets.
J Biol Chem
2009
, vol. 
284
 (pg. 
13413
-
13421
)
11
Worner
 
P
Brossmer
 
R
Platelet aggregation and the release induced by inophores for divalent cations.
Thromb Res
1975
, vol. 
6
 (pg. 
295
-
305
)
12
Feinstein
 
MB
Becker
 
EL
Fraser
 
C
Thrombin, collagen and A23187 stimulated endogenous platelet arachidonate metabolism: differential inhibition by PGE1, local anesthetics and a serine-protease inhibitor.
Prostaglandins
1977
, vol. 
14
 (pg. 
1075
-
1093
)
13
Feinstein
 
MB
Fraser
 
C
Human platelet secretion and aggregation induced by calcium ionophores: inhibition by PGE1 and dibutyryl cyclic AMP.
J Gen Physiol
1975
, vol. 
66
 (pg. 
561
-
581
)
14
Quinton
 
TM
Kim
 
S
Dangelmaier
 
C
, et al. 
Protein kinase C- and calcium-regulated pathways independently synergize with Gi pathways in agonist-induced fibrinogen receptor activation.
Biochem J
2002
, vol. 
368
 (pg. 
535
-
543
)
15
Quinton
 
TM
Ozdener
 
F
Dangelmaier
 
C
Daniel
 
JL
Kunapuli
 
SP
Glycoprotein VI-mediated platelet fibrinogen receptor activation occurs through calcium-sensitive and PKC-sensitive pathways without a requirement for secreted ADP.
Blood
2002
, vol. 
99
 (pg. 
3228
-
3234
)
16
Crittenden
 
JR
Bergmeier
 
W
Zhang
 
Y
, et al. 
CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation.
Nat Med
2004
, vol. 
10
 (pg. 
982
-
986
)
17
Cifuni
 
SM
Wagner
 
DD
Bergmeier
 
W
CalDAG-GEFI and protein kinase C represent alternative pathways leading to activation of integrin αIIbβ3 in platelets.
Blood
2008
, vol. 
112
 (pg. 
1696
-
1703
)
18
Kawasaki
 
H
Springett
 
GM
Toki
 
S
, et al. 
A Rap guanine nucleotide exchange factor enriched highly in the basal ganglia.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 (pg. 
13278
-
13283
)
19
Torti
 
M
Lapetina
 
EG
Structure and function of rap proteins in human platelets.
Thromb Haemostasis
1994
, vol. 
71
 (pg. 
533
-
543
)
20
Chrzanowska-Wodnicka
 
M
Smyth
 
SS
Schoenwaelder
 
SM
Fischer
 
TH
White
 
GC
Rap1b is required for normal platelet function and hemostasis in mice.
J Clin Invest
2005
, vol. 
115
 (pg. 
680
-
687
)
21
Caloca
 
MJ
Zugaza
 
JL
Bustelo
 
XR
Exchange factors of the RasGRP family mediate Ras activation in the Golgi.
J Biol Chem
2003
, vol. 
278
 (pg. 
33465
-
33473
)
22
Clyde-Smith
 
J
Silins
 
G
Gartside
 
M
, et al. 
Characterization of RasGRP2, a plasma membrane-targeted, dual specificity Ras/Rap exchange factor.
J Biol Chem
2000
, vol. 
275
 (pg. 
32260
-
32267
)
23
Bos
 
JL
Linking Rap to cell adhesion.
Curr Opin Cell Biol
2005
, vol. 
17
 (pg. 
123
-
128
)
24
York
 
RD
Yao
 
H
Dillon
 
T
, et al. 
Rap1 mediates sustained MAP kinase activation induced by nerve growth factor.
Nature
1998
, vol. 
392
 (pg. 
622
-
626
)
25
Stork
 
PJ
Dillon
 
TJ
Multiple roles of Rap1 in hematopoietic cells: complementary versus antagonistic functions.
Blood
2005
, vol. 
106
 (pg. 
2952
-
2961
)
26
Crittenden
 
JR
Cantuti-Castelvetri
 
I
Saka
 
E
, et al. 
Dysregulation of CalDAG-GEFI and CalDAG-GEFII predicts the severity of motor side-effects induced by anti-Parkinsonian therapy.
Proc Natl Acad Sci U S A
2009
, vol. 
106
 (pg. 
2892
-
2896
)
27
Guo
 
FF
Kumahara
 
E
Saffen
 
D
A CalDAG-GEFI/Rap1/B-Raf cassette couples M(1) muscarinic acetylcholine receptors to the activation of ERK1/2.
J Biol Chem
2001
, vol. 
276
 (pg. 
25568
-
25581
)
28
Lin
 
LL
Wartmann
 
M
Lin
 
AY
Knopf
 
JL
Seth
 
A
Davis
 
RJ
cPLA2 is phosphorylated and activated by MAP kinase.
Cell
1993
, vol. 
72
 (pg. 
269
-
278
)
29
Arita
 
H
Nakano
 
T
Hanasaki
 
K
Thromboxane A2: its generation and role in platelet activation.
Prog Lipid Res
1989
, vol. 
28
 (pg. 
273
-
301
)
30
Garcia
 
A
Quinton
 
TM
Dorsam
 
RT
Kunapuli
 
SP
Src family kinase-mediated and Erk-mediated thromboxane A2 generation are essential for VWF/GPIb-induced fibrinogen receptor activation in human platelets.
Blood
2005
, vol. 
106
 (pg. 
3410
-
3414
)
31
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
, vol. 
4
 (pg. 
638
-
647
)
32
Nadal-Wollbold
 
F
Pawlowski
 
M
Levy-Toledano
 
S
Berrou
 
E
Rosa
 
JP
Bryckaert
 
M
Platelet ERK2 activation by thrombin is dependent on calcium and conventional protein kinases C but not Raf-1 or B-Raf.
FEBS Lett
2002
, vol. 
531
 (pg. 
475
-
482
)
33
Bergmeier
 
W
Schulte
 
V
Brockhoff
 
G
Bier
 
U
Zirngibl
 
H
Nieswandt
 
B
Flow cytometric detection of activated mouse integrin αIIbβ3 with a novel monoclonal antibody.
Cytometry
2002
, vol. 
48
 (pg. 
80
-
86
)
34
Duncia
 
JV
Santella
 
JB
Higley
 
CA
, et al. 
MEK inhibitors: the chemistry and biological activity of U0126, its analogs, and cyclization products.
Bioorg Med Chem Lett
1998
, vol. 
8
 (pg. 
2839
-
2844
)
35
Guidetti
 
GF
Lova
 
P
Bernardi
 
B
, et al. 
The Gi-coupled P2Y12 receptor regulates diacylglycerol-mediated signaling in human platelets.
J Biol Chem
2008
, vol. 
283
 (pg. 
28795
-
28805
)
36
Hechler
 
B
Cattaneo
 
M
Gachet
 
C
The P2 receptors in platelet function.
Semin Thromb Hemostasis
2005
, vol. 
31
 (pg. 
150
-
161
)
37
Woulfe
 
D
Jiang
 
H
Mortensen
 
R
Yang
 
J
Brass
 
LF
Activation of Rap1B by Gi family members in platelets.
J Biol Chem
2002
, vol. 
277
 (pg. 
23382
-
23390
)
38
Eto
 
K
Murphy
 
R
Kerrigan
 
SW
, et al. 
Megakaryocytes derived from embryonic stem cells implicate CalDAG-GEFI in integrin signaling.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 (pg. 
12819
-
12824
)
39
Bertoni
 
A
Tadokoro
 
S
Eto
 
K
, et al. 
Relationships between Rap1b, affinity modulation of integrin αIIbβ3, and the actin cytoskeleton.
J Biol Chem
2002
, vol. 
277
 (pg. 
25715
-
25721
)
40
Watanabe
 
N
Bodin
 
L
Pandey
 
M
, et al. 
Mechanisms and consequences of agonist-induced talin recruitment to platelet integrin αIIbβ3.
J Cell Biol
2008
, vol. 
181
 (pg. 
1211
-
1222
)
41
Lee
 
HS
Lim
 
CJ
Puzon-McLaughlin
 
W
Shattil
 
SJ
Ginsberg
 
MH
RIAM activates integrins by linking talin to Ras GTPase membrane-targeting sequences.
J Biol Chem
2009
, vol. 
284
 (pg. 
5119
-
5127
)
42
Kitayama
 
H
Sugimoto
 
Y
Matsuzaki
 
T
Ikawa
 
Y
Noda
 
M
A ras-related gene with transformation suppressor activity.
Cell
1989
, vol. 
56
 (pg. 
77
-
84
)
43
Schmitt
 
JM
Stork
 
PJ
Cyclic AMP-mediated inhibition of cell growth requires the small G protein Rap1.
Mol Cell Biol
2001
, vol. 
21
 (pg. 
3671
-
3683
)
44
Vossler
 
MR
Yao
 
H
York
 
RD
Pan
 
MG
Rim
 
CS
Stork
 
PJ
cAMP activates MAP kinase and Elk-1 through a B-Raf- and Rap1-dependent pathway.
Cell
1997
, vol. 
89
 (pg. 
73
-
82
)
45
Ye
 
X
Shobe
 
JL
Sharma
 
SK
Marina
 
A
Carew
 
TJ
Small G proteins exhibit pattern sensitivity in MAPK activation during the induction of memory and synaptic facilitation in Aplysia.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 (pg. 
20511
-
20516
)
46
Garcia
 
J
de Gunzburg
 
J
Eychene
 
A
Gisselbrecht
 
S
Porteu
 
F
Thrombopoietin-mediated sustained activation of extracellular signal-regulated kinase in UT7-MpL cells requires both Ras-Raf-1- and Rap1-B-Raf-dependent pathways.
Mol Cell Biol
2001
, vol. 
21
 (pg. 
2659
-
2670
)
47
Ezumi
 
Y
Uchiyama
 
T
Takayama
 
H
Thrombopoietin potentiates the protein-kinase-C-mediated activation of mitogen-activated protein kinase/ERK kinases and extracellular signal-regulated kinases in human platelets.
Eur J Biochem
1998
, vol. 
258
 (pg. 
976
-
985
)
48
Kuijpers
 
MJ
Schulte
 
V
Bergmeier
 
W
, et al. 
Complementary roles of glycoprotein VI and α2β1 integrin in collagen-induced thrombus formation in flowing whole blood ex vivo.
FASEB J
2003
, vol. 
17
 (pg. 
685
-
687
)
49
Bernardi
 
B
Guidetti
 
GF
Campus
 
F
, et al. 
The small GTPase Rap1b regulates the cross talk between platelet integrin α2β1 and integrin αIIbβ3.
Blood
2006
, vol. 
107
 (pg. 
2728
-
2735
)
50
Kuijpers
 
MJ
Pozgajova
 
M
Cosemans
 
JM
, et al. 
Role of murine integrin α2β1 in thrombus stabilization and embolization: contribution of thromboxane A2.
Thromb Haemostasis
2007
, vol. 
98
 (pg. 
1072
-
1080
)
51
Bugaud
 
F
Nadal-Wollbold
 
F
Levy-Toledano
 
S
Rosa
 
JP
Bryckaert
 
M
Regulation of c-jun-NH2 terminal kinase and extracellular-signal regulated kinase in human platelets.
Blood
1999
, vol. 
94
 (pg. 
3800
-
3805
)
52
Kramer
 
RM
Roberts
 
EF
Strifler
 
BA
Johnstone
 
EM
Thrombin induces activation of p38 MAP kinase in human platelets.
J Biol Chem
1995
, vol. 
270
 (pg. 
27395
-
27398
)
53
Kauskot
 
A
Adam
 
F
Mazharian
 
A
, et al. 
Involvement of the mitogen-activated protein kinase c-Jun NH2-terminal kinase 1 in thrombus formation.
J Biol Chem
2007
, vol. 
282
 (pg. 
31990
-
31999
)
54
Borsch-Haubold
 
AG
Bartoli
 
F
Asselin
 
J
, et al. 
Identification of the phosphorylation sites of cytosolic phospholipase A2 in agonist-stimulated human platelets and HeLa cells.
J Biol Chem
1998
, vol. 
273
 (pg. 
4449
-
4458
)
55
Canobbio
 
I
Reineri
 
S
Sinigaglia
 
F
Balduini
 
C
Torti
 
M
A role for p38 MAP kinase in platelet activation by von Willebrand factor.
Thromb Haemostasis
2004
, vol. 
91
 (pg. 
102
-
110
)
56
Prevost
 
N
Mitsios
 
JV
Kato
 
H
, et al. 
Group IVA cytosolic phospholipase A2 (cPLA2α) and integrin αIIbβ3 reinforce each other's functions during αIIbβ3 signaling in platelets.
Blood
2009
, vol. 
113
 (pg. 
447
-
457
)
57
Cho
 
MJ
Liu
 
J
Pestina
 
TI
, et al. 
The roles of αIIbβ3-mediated outside-in signal transduction, thromboxane A2, and adenosine diphosphate in collagen-induced platelet aggregation.
Blood
2003
, vol. 
101
 (pg. 
2646
-
2651
)
58
Cowan
 
DH
Platelet adherence to collagen: role of prostaglandin-thromboxane synthesis.
Br J Haematol
1981
, vol. 
49
 (pg. 
425
-
434
)
59
Michelson
 
AD
P2Y12 antagonism: promises and challenges.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 (pg. 
s33
-
s38
)
60
Schultess
 
J
Danielewski
 
O
Smolenski
 
AP
Rap1GAP2 is a new GTPase-activating protein of Rap1 expressed in human platelets.
Blood
2005
, vol. 
105
 (pg. 
3185
-
3192
)
61
Lorenowicz
 
MJ
van Gils
 
J
de Boer
 
M
Hordijk
 
PL
Fernandez-Borja
 
M
Epac1-Rap1 signaling regulates monocyte adhesion and chemotaxis.
J Leukocyte Biol
2006
, vol. 
80
 (pg. 
1542
-
1552
)
62
He
 
JC
Neves
 
SR
Jordan
 
JD
Iyengar
 
R
Role of the Go/i signaling network in the regulation of neurite outgrowth.
Can J Physiol Pharmacol
2006
, vol. 
84
 (pg. 
687
-
694
)
Sign in via your Institution