Background/Aims: DAPT (24-diamino-5-phenylthiazole) inhibits γ-secretase, which cleaves the signaling molecule CD44, a negative regulator of platelet activation and apoptosis. CD44 is a co-receptor for macrophage migration inhibitory factor (MIF) an anti-apoptotic pro-inflammatory cytokine expressed and released from blood platelets. Whether DAPT influences platelet function, remained, however, elusive. Activators of platelets include collagen related peptide (CRP). The present study thus explored whether DAPT modifies the stimulating effect of CRP on platelet function. Methods: Platelets isolated from wild-type mice were exposed for 30 minutes to DAPT (10 µM). Flow cytometry was employed to estimate Orai1 abundance with specific antibodies, cytosolic Ca2+-activity ([Ca2+]i) from Fluo-3 fluorescence, platelet degranulation from P-selectin abundance, integrin activation from αIIbβ3 integrin abundance, generation of reactive oxygen species (ROS) from DCFDA fluorescence, mitochondrial transmembrane potential from TMRE fluorescence, phospholipid scrambling of the cell membrane from annexin-V-binding, relative platelet volume from forward scatter and aggregation utilizing staining with CD9-APC and CD9-PE. Results: Exposure of platelets to 2-5 µg/ml CRP was followed by significant increase of Orai1 abundance, [Ca2+]i, and P-selectin abundance, as well as by αIIbβ3 integrin activation, ROS generation, mitochondrial depolarization, enhanced annexin-V-binding, decreased cell volume, and aggregation. All CRP induced effects were significantly blunted in the presence of DAPT. Conclusions: The γ-secretase inhibitor DAPT counteracts agonist induced platelet activation, apoptosis and aggregation.

DAPT (24-diamino-5-phenylthiazole) [1] is a known inhibitor of γ-secretase [2,3]. The enzyme cleaves the signaling molecule CD44 [4,5,6], a multifunctional transmembrane glycoprotein and (co)receptor [7,8] expressed in a wide variety of normal and cancer cells [7,9] and contributing to the regulation of diverse cellular functions including cell adhesion, proliferation, growth, survival, motility, migration, angiogenesis, and differentiation [7,8,10,11,12,13].

CD44 is involved in the signaling of macrophage migration inhibitory factor (MIF) [14,15], which is expressed and released from blood platelets [16]. Platelets are required for primary hemostasis following vascular injury and contribute to the pathophysiology of acute thrombotic occlusion following atherosclerotic plaque rupture [17,18]. Platelets are involved in the pathophysiology of arterial thrombosis, vascular inflammation and atherogenesis [17,19]. They are activated by an increase of cytosolic Ca2+ concentration ([Ca2+]i) [20], resulting from Ca2+ release from intracellular stores [21] with subsequent activation of the Ca2+ release-activated channel (CRAC) Orai1 (CRACM1) in the plasma membrane [20,22,23,24]. Most recently CD44 has been disclosed as a negative regulator of platelet activation [25]. Specifically, CD44 deficiency has been shown to augment in platelets thrombin and collagen related peptide induced increase of Orai1 surface abundance, [Ca2+]i, degranulation, αIIbβ3 integrin activation, caspase-3 activity, cell membrane scrambling, and cell shrinkage. Moreover, CD44 deficiency augmented platelet adhesion and in vitro thrombus formation under high arterial shear rates [25].

Inhibitors of γ-secretase were expected to enhance the abundance of CD44 with the respective down-regulation of platelet activation and apoptosis.

Nothing is known about any potential effects of DAPT on platelet function. The present study thus explored whether DAPT modifies the effect of collagen related peptide on platelet activation with Ca2+ entry and phospholipid scrambling of the cell membrane.

Mice

All animal experiments were conducted according to the German law for the welfare of animals and were approved by the authorities of the state of Baden-Württemberg. Experiments were performed with blood platelets isolated from wild type mice. The mice had free access to water and control chow (Ssniff, Soest, Germany).

Preparation of mouse platelets

Platelets were obtained from 10- to 12-week-old mice of either sex. The mice were anesthetized and 800 µl blood was drawn from the retro-orbital plexus into tubes with 200 µl acid-citrate-dextrose buffer before the mice were sacrificed [26]. Platelet rich plasma (PRP) was obtained by centrifugation at 260 g for 5 minutes. Afterwards PRP was centrifuged at 640 g for 5 minutes to pellet the platelets. Where necessary apyrase (0.02 U/ml; Sigma-Aldrich) and prostaglandin I2 (0.5 µM; Calbiochem) were added to the PRP to prevent activation of platelets during isolation. After two washing steps the pellet of washed platelets was resuspended in modified Tyrode-HEPES buffer (pH 7.4, supplemented with 1 mM CaCl2). Where indicated, CRP (kindly provided by R.Farndale, University of Cambridge, Cambridge, UK) was added.

Orai1 surface abundance

Orai1 surface expression was analyzed in platelets by flow cytometry. Washed platelets were incubated with 2 µg/ml CRP without BSA, and subsequently fixed with 1% paraformaldehyde for 30 minutes on ice. After rinsing three times, platelets were incubated for 90 minutes with Orai1 rabbit anti-mouse antibody (Abcam), washed once in Tyrode buffer, and stained in 1:250 diluted CF™ 488A-labeled anti-rabbit secondary antibody (Sigma, USA) for 60 minutes. Samples were immediately analyzed on a FACS Calibur flow cytometer (BD Biosciences).

Cytosolic calcium

For the measurement of the cytosolic Ca2+ concentration the platelet preparation was washed once in Tyrode buffer (pH 7.4), stained with 3 µM Fluo-3AM (Biotinium, USA) in the same buffer and incubated at 37°C for 30 minutes. Following the indicated experimental treatment, relative fluorescence was measured utilizing a BD FACS Calibur (BD Biosciences, Heidelberg, Germany) [27].

P-selectin and activated integrin abundance

Fluorophore-labeled antibodies were utilized for the detection of P-selectin expression (Wug.E9-FITC) and the active form of αIIbβ3 integrin (JON/A-PE). Washed mouse platelets (1x106) were suspended in modified Tyrode buffer (pH 7.4) containing 1 mM CaCl2 and antibodies (1:10 dilution) and subsequently subjected to the respective treatments and for the indicated time periods at room temperature (RT). The reaction was stopped by addition of PBS and the samples were immediately analyzed on a BD FACSCalibur.

Reactive oxygen species

The abundance of reactive oxygen species (ROS) was determined utilizing 2′,7′-dichlorodihydrofluorescein diacetate (DCFDA). To load the platelets, DCFDA (Sigma, Schnelldorf, Germany) was added to the cell suspension at a final concentration of 10 µM. Following the indicated experimental treatments, fluorescence was measured by flow cytometry utilizing a BD FACSCalibur (BD Biosciences, Heidelberg, Germany).

Mitochondrial Membrane Potential (Δψm)

Mitochondrial membrane potential was assessed by flow cytometry [28]. Briefly, platelets were incubated in 10 µM DAPT for 30 min, and then treated with 5 µg/ml collagen related peptide for 60 minutes. Pre-incubated platelets were stained with 10 µM of TMRE (Invitrogen, USA) for 30 minutes at room temperature in the dark, and the TMRE-fluorescence was measured by flow cytometry utilizing a BD FACSCalibur.

Phosphatidylserine exposure and forward scatter

Phosphatidylserine exposure was determined in platelets with and without 10 minutes CRP treatment. To this end, the platelet preparation was centrifuged at 660 g for 5 minutes followed by washing once with Tyrode buffer (pH 7.4) with 1 mM CaCl2, staining with 1:20 dilution of Annexin-V FITC (Mabtag, Germany) in Tyrode buffer (pH 7.4) with 2 mM CaCl2 and incubation at 37°C for 30 minutes. Annexin-V binding reflecting surface exposure of phosphatidylserine was evaluated by flow cytometry utilizing a BD FACSCalibur. In parallel, the forward scatter (FSC) of the platelets was determined by flow cytometry as a measure of platelet size.

Platelet aggregation

Aggregation was determined utilizing flow cytometry as previously described [29]. To this end platelets were labeled with CD9-APC and CD9-PE monoclonal antibodies (1:100 dilution, Abcam) for 15 minutes at room temperature. Following incubation, differently labeled samples were washed twice, mixed 1:1, and incubated in 10 µM DAPT (Sigma, Germany) for 30 min at 37°C while shaking at 600 rpm for 10 minutes. Pre-incubated platelets were activated with 2 µg/ml collagen related peptide at 37°C while shaking at 1000 rpm. At the indicated time points, samples were fixed by addition 0.5% paraformaldehyde (Carl Roth, Germany) in phosphate-buffered saline. The fixed samples were measured utilizing a BD FACSCalibur (BD Biosciences, Heidelberg, Germany). For quantification, a quadrant was set in the dot plot of respective channels on non-stimulated platelets. The appearance of double-colored events in the upper right quadrant (Q2) was quantified as percentage of total amount of labeled events (Q1+Q2+Q4) at every time point analyzed.

Statistical analysis

Data are provided as means ± SEM; n represents the number of independent experiments. All data were tested for significance using ANOVA with Tukey's test as post-test or unpaired student's t-test as appropriate. Results with p<0.05 were considered statistically significant.

The present study explored whether γ-secretase inhibitor DAPT (24-diamino-5-phenylthiazole) influences platelet activation. To this end, murine platelets were isolated from wild type mice and activated with collagen related peptide (CRP).

In a first series of experiments the abundance of Orai1 protein in the platelet cell membrane was quantified. As illustrated in Fig. 1A and D, without CRP treatment Orai1 protein expression in the platelet membrane was similarly low in presence and absence of γ-secretase inhibitor DAPT (10 µM). Activation of platelets with 2 µg/ml CRP was followed by a significant increase of Orai1 protein abundance. After short time treatment (100 seconds) Orai1 protein abundance was not significantly different in presence or absence of DAPT (Fig. 1B, D). After 15 minutes CRP treatment, the increase of Orai1 abundance in the platelet plasma membrane was, however, significantly blunted in the presence of 10 µM DAPT (Fig. 1C, D).

Fig. 1

DAPT and CRP sensitive Orai1 protein abundance in the cell membrane. A,B,C. Original histogram overlays of Orai1 protein abundance in platelets isolated from mice without (grey shadows) and with (black lines) presence of 10 µM DAPT prior to (A) and following a 100 sec (B) or 15 min (C) treatment with 2 µg/ml collagen related peptide. D. Arithmetic means ± SEM (n = 5) of the anti-Orai1 fluorescence (arbitrary fluorescence units) in murine platelets without (white bars) and with (black bars) the presence of 10 µM γ-secretase inhibitor DAPT prior to (left bars) and following a 100 sec (middle bars) or 15 min (right bars) treatment with collagen related peptide (CRP, 2 µg/ml). E. Arithmetic means ± SEM (n = 5) of the anti-Orai1 fluorescence (arbitrary fluorescence units) in murine platelets without (white bars) and with (black bars) the presence of 1, 5, 10 or 20 µM γ-secretase inhibitor DAPT prior to (left bars) and following (right bars) a 100 sec treatment with collagen related peptide (CRP, 2 µg/ml). ###(p<0.001) indicates statistically significant difference from absence of CRP, *(p<0.05) and **(p<0.01) indicate statistically significant difference from absence of DAPT.

Fig. 1

DAPT and CRP sensitive Orai1 protein abundance in the cell membrane. A,B,C. Original histogram overlays of Orai1 protein abundance in platelets isolated from mice without (grey shadows) and with (black lines) presence of 10 µM DAPT prior to (A) and following a 100 sec (B) or 15 min (C) treatment with 2 µg/ml collagen related peptide. D. Arithmetic means ± SEM (n = 5) of the anti-Orai1 fluorescence (arbitrary fluorescence units) in murine platelets without (white bars) and with (black bars) the presence of 10 µM γ-secretase inhibitor DAPT prior to (left bars) and following a 100 sec (middle bars) or 15 min (right bars) treatment with collagen related peptide (CRP, 2 µg/ml). E. Arithmetic means ± SEM (n = 5) of the anti-Orai1 fluorescence (arbitrary fluorescence units) in murine platelets without (white bars) and with (black bars) the presence of 1, 5, 10 or 20 µM γ-secretase inhibitor DAPT prior to (left bars) and following (right bars) a 100 sec treatment with collagen related peptide (CRP, 2 µg/ml). ###(p<0.001) indicates statistically significant difference from absence of CRP, *(p<0.05) and **(p<0.01) indicate statistically significant difference from absence of DAPT.

Close modal

In order to test, whether DAPT modified cytosolic Ca2+ concentration ([Ca2+]i) in murine platelets, [Ca2+]i was determined utilizing Fluo-3 fluorescence. As illustrated in Fig. 2A and C, prior to CRP treatment [Ca2+]i was similar in the absence and presence of DAPT. Treatment with CRP (2 µg/ml) was within 100 seconds followed by a significant increase of [Ca2+]i in platelets, an effect significantly blunted in the presence of DAPT (Fig. 2B, C).

Fig. 2

DAPT sensitive CRP-induced increase of cytosolic Ca2+ concentration. A,B. Original histogram overlays of Fluo-3 fluorescence reflecting cytosolic Ca2+ activity in murine platelets without (A) and with (B) a 100 sec treatment with CRP (2 µg/ml). C,D. Arithmetic means ± SEM (n = 4) of Fluo-3 fluorescence reflecting cytosolic Ca2+ activity in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) CRP treatment. Shown is (C) the effect of 2 µg/ml CRP at different exposure times and (D) the effect of different CRP concentrations after 100 seconds exposure. ###(p<0.001) indicates statistically significant difference from absence of CRP, **(p<0.01) and *** (p<0.001)indicate statistically significant difference from absence of DAPT.

Fig. 2

DAPT sensitive CRP-induced increase of cytosolic Ca2+ concentration. A,B. Original histogram overlays of Fluo-3 fluorescence reflecting cytosolic Ca2+ activity in murine platelets without (A) and with (B) a 100 sec treatment with CRP (2 µg/ml). C,D. Arithmetic means ± SEM (n = 4) of Fluo-3 fluorescence reflecting cytosolic Ca2+ activity in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) CRP treatment. Shown is (C) the effect of 2 µg/ml CRP at different exposure times and (D) the effect of different CRP concentrations after 100 seconds exposure. ###(p<0.001) indicates statistically significant difference from absence of CRP, **(p<0.01) and *** (p<0.001)indicate statistically significant difference from absence of DAPT.

Close modal

Platelet degranulation was estimated from the increase of P-selectin abundance on the platelet surface, which was determined utilizing specific antibodies and flow cytometry. As illustrated in Fig. 3A and C, the P-selectin abundance was negligible at the surface of resting platelets and not significantly modified by DAPT treatment. CRP (2 µg/ml) treatment was followed by a sharp increase of P-selectin abundance, an effect significantly blunted in the presence of 10 µM DAPT (Fig. 3B, C).

Fig. 3

DAPT sensitive CRP-induced platelet degranulation and integrin αIIbβ3 activation. A,B. Original histogram overlays of P-selectin related fluorescence in murine platelets without (A) and with (B) a 15 min CRP (2 µg/ml) treatment. C. Arithmetic means ± SEM (n = 4) of the P-selectin related fluorescence (arbitrary units) in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 15 min CRP treatment (2 µg/ml). D,E. Original histogram overlays of activated αIIbβ3 integrin related fluorescence in murine platelets without (D) and with (E) a 15 min CRP (2 µg/ml) treatment. F. Arithmetic means ± SEM (n = 4) of activated αIIbβ3 integrin related fluorescence (arbitrary units) in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 15 min CRP treatment (2 µg/ml). ###(p<0.001) indicates statistically significant difference from absence of CRP, *(p<0.05) indicate statistically significant difference from absence of DAPT.

Fig. 3

DAPT sensitive CRP-induced platelet degranulation and integrin αIIbβ3 activation. A,B. Original histogram overlays of P-selectin related fluorescence in murine platelets without (A) and with (B) a 15 min CRP (2 µg/ml) treatment. C. Arithmetic means ± SEM (n = 4) of the P-selectin related fluorescence (arbitrary units) in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 15 min CRP treatment (2 µg/ml). D,E. Original histogram overlays of activated αIIbβ3 integrin related fluorescence in murine platelets without (D) and with (E) a 15 min CRP (2 µg/ml) treatment. F. Arithmetic means ± SEM (n = 4) of activated αIIbβ3 integrin related fluorescence (arbitrary units) in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 15 min CRP treatment (2 µg/ml). ###(p<0.001) indicates statistically significant difference from absence of CRP, *(p<0.05) indicate statistically significant difference from absence of DAPT.

Close modal

Similarly, the abundance of active integrin αIIbβ3 was negligible at the surface of resting platelets (Fig. 3D, F) and was significantly increased by treatment with CRP (2 µg/ml). Again, the effect was significantly blunted in the presence of 10 µM DAPT (Fig. 3E, F).

Further experiments were performed to test, whether DAPT modifies the abundance of reactive oxygen species (ROS). ROS generation was quantified utilizing DCFDA fluorescence. As shown in Fig. 4, following a 2 µg/ml CRP treatment, ROS generation was increased, an effect significantly blunted in the presence of DAPT after 7 minutes CRP treatment.

Fig. 4

DAPT sensitive CRP-induced increase of reactive oxygen species. Arithmetic means ± SEM (n = 4) of DCFDA fluorescence reflecting ROS abundance (arbitrary units) in murine platelets in the absence (black circles) and presence (black squares) of DAPT following a 2 µg/ml CRP treatment. *(p<0.05) indicate statistically significant difference from absence of DAPT.

Fig. 4

DAPT sensitive CRP-induced increase of reactive oxygen species. Arithmetic means ± SEM (n = 4) of DCFDA fluorescence reflecting ROS abundance (arbitrary units) in murine platelets in the absence (black circles) and presence (black squares) of DAPT following a 2 µg/ml CRP treatment. *(p<0.05) indicate statistically significant difference from absence of DAPT.

Close modal

In a further series of experiments the effect of DAPT and CRP on mitochondrial membrane potential (Δψm) was determined with TMRE by flow cytometry. As illustrated in Fig. 5A and C, Δψm was similar in resting platelets both, with or without presence of DAPT. However, TMRE fluorescence was decreased by treatment with 5 µg/ml CRP, an effect again significantly less pronounced in the presence than in the absence of DAPT (Fig. 5B, C).

Fig. 5

DAPT sensitive CRP-induced mitochondrial depolarization. A,B. Original histogram overlays of TMRE fluorescence reflecting Δψm in murine platelets without (A) and with (B) 5 µg/ml CRP treatment. C. Arithmetic means ± SEM (n = 5) of TMRE fluorescence reflecting Δψm (arbitrary units) in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) CRP treatment. ###(p<0.001) indicates statistically significant difference from absence of CRP, **(p<0.01) indicate statistically significant difference from absence of DAPT

Fig. 5

DAPT sensitive CRP-induced mitochondrial depolarization. A,B. Original histogram overlays of TMRE fluorescence reflecting Δψm in murine platelets without (A) and with (B) 5 µg/ml CRP treatment. C. Arithmetic means ± SEM (n = 5) of TMRE fluorescence reflecting Δψm (arbitrary units) in murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) CRP treatment. ###(p<0.001) indicates statistically significant difference from absence of CRP, **(p<0.01) indicate statistically significant difference from absence of DAPT

Close modal

As illustrated in Fig. 6A and C, the percentage of annexin-V positive platelets was again negligible in untreated platelets, irrespective of the presence of DAPT. CRP (5 µg/ml) within 10 min significantly enhanced the percentage of annexin-V binding platelets, an effect again significantly blunted in the presence of DAPT (10 µM) (Fig. 6B, C).

Fig. 6

DAPT sensitive CRP-dependent cell membrane scrambling and cell volume. A,B. Original histogram overlays of annexin-V-binding reflecting phosphatidylserine abundance at the surface of murine platelets without (A) and with (B) a 10 min treatment with CRP (5 µg/ml). C. Arithmetic means ± SEM (n = 4) of the percentage of annexin-V-binding murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 10 min CRP treatment (5 µg/ml). D,E. Original histogram overlays of forward scatter reflecting cell volume of murine platelets without (D) and with (E) a 10 min treatment with CRP (5 µg/ml). F. Arithmetic means ± SEM (n = 4) of forward scatter reflecting cell volume of murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 10 min CRP treatment (5 µg/ml). ###(p<0.001) indicates statistically significant difference from absence of CRP, *(p<0.05) indicate statistically significant difference from absence of DAPT.

Fig. 6

DAPT sensitive CRP-dependent cell membrane scrambling and cell volume. A,B. Original histogram overlays of annexin-V-binding reflecting phosphatidylserine abundance at the surface of murine platelets without (A) and with (B) a 10 min treatment with CRP (5 µg/ml). C. Arithmetic means ± SEM (n = 4) of the percentage of annexin-V-binding murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 10 min CRP treatment (5 µg/ml). D,E. Original histogram overlays of forward scatter reflecting cell volume of murine platelets without (D) and with (E) a 10 min treatment with CRP (5 µg/ml). F. Arithmetic means ± SEM (n = 4) of forward scatter reflecting cell volume of murine platelets in the absence (white bars) and presence (black bars) of DAPT without (left bars) and with (right bars) a 10 min CRP treatment (5 µg/ml). ###(p<0.001) indicates statistically significant difference from absence of CRP, *(p<0.05) indicate statistically significant difference from absence of DAPT.

Close modal

Platelet volume was estimated from forward scatter, which was determined by flow cytometry. As illustrated in Fig. 6D and F, prior to stimulation with CRP, forward scatter was again similar in the absence and presence of DAPT. CRP (5 µg/ml) treatment within 10 min was followed by a significant decrease of forward scatter, an effect significantly less pronounced in the presence than in the absence of DAPT (Fig. 6E, F).

To elucidate the effect of DAPT and CRP on platelet aggregation, platelets were labeled with two distinct dyes and the coincidence of the two dyes estimated by flow cytometry. As illustrated in Fig. 7, aggregation of resting platelets was similarly low in DAPT treated and untreated platelets. CRP (2 µg/ml) treatment within a few min significantly increased the platelet aggregation, an effect significantly blunted by prior DAPT treatment.

Fig. 7

DAPT sensitive CRP-induced platelet aggregation. A. Original dot blots reflecting platelet aggregation without (a,c) and with (b,d) prior DAPT (10 µM) treatment, and subsequent treatment with CRP for 0 min (a,b) and 4 min (c,d). B. Arithmetic means ± SEM (n = 4) of platelet aggregation without (black circles) and with (black squares) prior DAPT (10 µM) treatment as a function of time after addition of CRP (2 µg/ml). *(p<0.05) indicate statistically significant difference from absence of DAPT.

Fig. 7

DAPT sensitive CRP-induced platelet aggregation. A. Original dot blots reflecting platelet aggregation without (a,c) and with (b,d) prior DAPT (10 µM) treatment, and subsequent treatment with CRP for 0 min (a,b) and 4 min (c,d). B. Arithmetic means ± SEM (n = 4) of platelet aggregation without (black circles) and with (black squares) prior DAPT (10 µM) treatment as a function of time after addition of CRP (2 µg/ml). *(p<0.05) indicate statistically significant difference from absence of DAPT.

Close modal

The present observations disclose a novel effect of the γ-secretase inhibitor DAPT (24-diamino-5-phenylthiazole), i.e. the attenuation of platelet activation following treatment with collagen related peptide (CRP). In the absence of CRP, treatment of platelets with DAPT had little effect on the tested platelet properties. In contrast, DAPT significantly blunted the effect of CRP on degranulation, integrin activation, cell membrane scrambling and cell shrinkage. Phosphatidylserine translocation to the platelet surface following cell membrane scrambling is expected to foster the procoagulant function of platelets and is thus decisive in hemostasis [30].

The effects of DAPT could be explained by inhibition of γ-secretase which would be expected to enhance the CD44 protein abundance and thus augment the inhibitory effect of CD44 on platelet activation and apoptosis [25].

Activation of platelets with thrombus formation and apoptosis is in large part secondary to increase of cytosolic Ca2+ activity ([Ca2+]i) [23,31] and the negative effects of DAPT on activation and cell membrane scrambling is paralleled by a significantly blunted increase of [Ca2+]i. The increase of [Ca2+]i following CRP treatment could be contributing to oxidative stress. Along those lines, DAPT blunted the increase of reactive oxygen species (ROS) following CRP treatment. The oxidative stress is paralleled by mitochondrial depolarization. CD44 counteracts oxidative stress [32] and could thus contribute to or even account for the attenuation of ROS formation by DAPT.

An increase of [Ca2+]i leads to platelet activation, which fosters the development of arterial thrombosis [20]. An increase of [Ca2+]i further triggers phospholipid scrambling of the cell membrane with translocation of phosphatidylserine to the platelet surface [19,33,34,35]. The inhibition of CRP-induced phosphatidylserine exposure may thus result from a decrease of Ca2+ entry. Phosphatidylserine exposed at the platelet surface stimulates thrombin formation, which in turn augments the activation of platelets [33,34,35,36]. Phosphatidylserine exposing platelets are further bound to and phagocytosed by macrophages [37]. Both, platelet activation and apoptosis contribute to the stimulation of platelet aggregation, which is again slightly, but significantly blunted by DAPT.

Upon stimulation, platelets release the CD44 activating ligand macrophage migration inhibitory factor (MIF) [16], which inhibits, as part of an autocrine negative feedback loop, platelet cell membrane scrambling. MIF limits activation-induced apoptosis via interaction with CXCR7 [28]. CD44 is considered a target in the treatment of inflammatory disease [15]. To the extent that γ-secretase degrades CD44 [4,5,6], pharmacological inhibition γ-secretase could strengthen the negative feedback loop and thus down-regulate platelet activation and phospholipid scrambling of the platelet plasma membrane. However, even though CD44 participates in the signaling of MIF [14,15], and lack of CD44 augments the activation of platelets by several agonists such as CRP [25], the observed blunting effect of DAPT on platelet activation and cell membrane scrambling is not necessarily due to increased CD44 abundance and activity.

In conclusion, the γ-secretase inhibitor DAPT attenuates the CRP-induced increase of Orai1 abundance, [Ca2+]i, oxidative stress, degranulation, integrin activation, translocation of phosphatidylserine to the platelet cell membrane surface and platelet shrinkage, and thus attenuates platelet activation and apoptosis.

We thank Efi Faber for providing technical assistance as well as Tanja Loch for meticulous preparation of the manuscript. This study was supported by the Deutsche Forschungsgemeinschaft - Klinische Forschergruppe [DFG-KFO 274] ‘Platelets—Molecular Mechanisms and Translational Implications', as well as the Tuebingen Platelet Investigative Consortium (TuePIC), and the Open Access Publishing Fund of Tuebingen University.

The authors of this manuscript state that they have no conflicts of interest to declare.

1.
Liu Y, Li P, Liu K, He Q, Han S, Sun X, Li T, Shen L: Timely inhibition of Notch signaling by DAPT promotes cardiac differentiation of murine pluripotent stem cells. PLoS One 2014;9:e109588.
2.
Jiang LY, Zhang XL, Du P, Zheng JH: gamma-Secretase Inhibitor, DAPT Inhibits Self-renewal and Stemness Maintenance of Ovarian Cancer Stem-like Cells In Vitro. Chin J Cancer Res 2011;23:140-146.
3.
Li LC, Peng Y, Liu YM, Wang LL, Wu XL: Gastric cancer cell growth and epithelial-mesenchymal transition are inhibited by gamma-secretase inhibitor DAPT. Oncol Lett 2014;7:2160-2164.
4.
McManus MM, Weiss KR, Hughes DP: Understanding the role of Notch in osteosarcoma. Adv Exp Med Biol 2014;804:67-92.
5.
Nagase H, Koh CS, Nakayama K: gamma-Secretase-regulated signaling pathways, such as notch signaling, mediate the differentiation of hematopoietic stem cells, development of the immune system, and peripheral immune responses. Curr Stem Cell Res Ther 2011;6:131-141.
6.
Nagase H, Nakayama K: gamma-Secretase-regulated signaling typified by Notch signaling in the immune system. Curr Stem Cell Res Ther 2013;8:341-356.
7.
Jaggupilli A, Elkord E: Significance of CD44 and CD24 as cancer stem cell markers: an enduring ambiguity. Clin Dev Immunol 2012;2012:708036.
8.
Naor D, Wallach-Dayan SB, Zahalka MA, Sionov RV: Involvement of CD44, a molecule with a thousand faces, in cancer dissemination. Semin Cancer Biol 2008;18:260-267.
9.
Ponta H, Sherman L, Herrlich PA: CD44: from adhesion molecules to signalling regulators. Nat Rev Mol Cell Biol 2003;4:33-45.
10.
Bourguignon LY: CD44-mediated oncogenic signaling and cytoskeleton activation during mammary tumor progression. J Mammary Gland Biol Neoplasia 2001;6:287-297.
11.
Du L, Wang H, He L, Zhang J, Ni B, Wang X, Jin H, Cahuzac N, Mehrpour M, Lu Y, Chen Q: CD44 is of functional importance for colorectal cancer stem cells. Clin Cancer Res 2008;14:6751-6760.
12.
Napier SL, Healy ZR, Schnaar RL, Konstantopoulos K: Selectin ligand expression regulates the initial vascular interactions of colon carcinoma cells: the roles of CD44v and alternative sialofucosylated selectin ligands. J Biol Chem 2007;282:3433-3441.
13.
Rangaswami H, Bulbule A, Kundu GC: Osteopontin: role in cell signaling and cancer progression. Trends Cell Biol 2006;16:79-87.
14.
Merk M, Mitchell RA, Endres S, Bucala R: D-dopachrome tautomerase (D-DT or MIF-2): doubling the MIF cytokine family. Cytokine 2012;59:10-17.
15.
Sanchez-Nino MD, Sanz AB, Ruiz-Andres O, Poveda J, Izquierdo MC, Selgas R, Egido J, Ortiz A: MIF, CD74 and other partners in kidney disease: tales of a promiscuous couple. Cytokine Growth Factor Rev 2013;24:23-40.
16.
Strussmann T, Tillmann S, Wirtz T, Bucala R, von Hundelshausen P, Bernhagen J: Platelets are a previously unrecognised source of MIF. Thromb Haemost 2013;110:1004-1013.
17.
Gawaz M: Role of platelets in coronary thrombosis and reperfusion of ischemic myocardium. Cardiovasc Res 2004;61:498-511.
18.
Borst O, Schmidt EM, Munzer P, Schonberger T, Towhid ST, Elvers M, Leibrock C, Schmid E, Eylenstein A, Kuhl D, May AE, Gawaz M, Lang F: The serum- and glucocorticoid-inducible kinase 1 (SGK1) influences platelet calcium signaling and function by regulation of Orai1 expression in megakaryocytes. Blood 2012;119:251-261.
19.
Borst O, Munzer P, Gatidis S, Schmidt EM, Schonberger T, Schmid E, Towhid ST, Stellos K, Seizer P, May AE, Lang F, Gawaz M: The inflammatory chemokine CXC motif ligand 16 triggers platelet activation and adhesion via CXC motif receptor 6-dependent phosphatidylinositide 3-kinase/Akt signaling. Circ Res 2012;111:1297-1307.
20.
Bergmeier W, Stefanini L: Novel molecules in calcium signaling in platelets. J Thromb Haemost 2009;7:S187-190.
21.
Varga-Szabo D, Braun A, Nieswandt B: Calcium signaling in platelets. J Thromb Haemost 2009;7:1057-1066.
22.
Braun A, Varga-Szabo D, Kleinschnitz C, Pleines I, Bender M, Austinat M, Bosl M, Stoll G, Nieswandt B: Orai1 (CRACM1) is the platelet SOC channel and essential for pathological thrombus formation. Blood 2009;113:2056-2063.
23.
Varga-Szabo D, Braun A, Nieswandt B: STIM and Orai in platelet function. Cell Calcium 2011;50:270-278.
24.
Wang Y, Deng X, Gill DL: Calcium signaling by STIM and Orai: intimate coupling details revealed. Sci Signal 2010;3:e42.
25.
Liu G, Liu G, Alzoubi K, Chatterjee M, Walker B, Munzer P, Luo D, Umbach AT, Elvira B, Chen H, Voelkl J, Foller M, Mak TW, Borst O, Gawaz M, Lang F: CD44 sensitivity of platelet activation, membrane scrambling and adhesion under high arterial shear rates. Thromb Haemost 2015;115:99-108.
26.
Pelzl L, Fakhri H, Umbach AT, Gawaz M, Paulmichl M, Lang F: Sgk1 sensitive pendrin expression in murine platelets. Cell Physiol Biochem 2013;32:210-220.
27.
Towhid ST, Schmidt EM, Tolios A, Munzer P, Schmid E, Borst O, Gawaz M, Stegmann E, Lang F: Stimulation of platelet death by vancomycin. Cell Physiol Biochem 2013;31:102-112.
28.
Chatterjee M, Borst O, Walker B, Fotinos A, Vogel S, Seizer P, Mack A, Alampour-Rajabi S, Rath D, Geisler T, Lang F, Langer HF, Bernhagen J, Gawaz M: Macrophage migration inhibitory factor limits activation-induced apoptosis of platelets via CXCR7-dependent Akt signaling. Circ Res 2014;115:939-949.
29.
De Cuyper IM, Meinders M, van de Vijver E, de Korte D, Porcelijn L, de Haas M, Eble JA, Seeger K, Rutella S, Pagliara D, Kuijpers TW, Verhoeven AJ, van den Berg TK, Gutierrez L: A novel flow cytometry-based platelet aggregation assay. Blood 2013;121:e70-80.
30.
Lhermusier T, Chap H, Payrastre B: Platelet membrane phospholipid asymmetry: from the characterization of a scramblase activity to the identification of an essential protein mutated in Scott syndrome. J Thromb Haemost 2011;9:1883-1891.
31.
Lang F, Munzer P, Gawaz M, Borst O: Regulation of STIM1/Orai1-dependent Ca2+ signalling in platelets. Thromb Haemost 2013;110:925-930.
32.
Yoshida GJ, Saya H: Inversed relationship between CD44 variant and c-Myc due to oxidative stress-induced canonical Wnt activation. Biochem Biophys Res Commun 2014;443:622-627.
33.
Harper MT, Poole AW: Store-operated calcium entry and non-capacitative calcium entry have distinct roles in thrombin-induced calcium signalling in human platelets. Cell Calcium 2011;50:351-358.
34.
Mahaut-Smith MP: A role for platelet TRPC channels in the Ca2+ response that induces procoagulant activity. Sci Signal 2013;6:pe23.
35.
Mushtaq M, Nam TS, Kim UH: Critical role for CD38-mediated Ca2+ signaling in thrombin-induced procoagulant activity of mouse platelets and hemostasis. J Biol Chem 2011;286:12952-12958.
36.
Wolfs JL, Comfurius P, Rasmussen JT, Keuren JF, Lindhout T, Zwaal RF, Bevers EM: Activated scramblase and inhibited aminophospholipid translocase cause phosphatidylserine exposure in a distinct platelet fraction. Cell Mol Life Sci 2005;62:1514-1525.
37.
Badlou BA, Wu YP, Smid WM, Akkerman JW: Platelet binding and phagocytosis by macrophages. Transfusion 2006;46:1432-1443.

G. Liu and G. Liu contributed equally and thus share first authorship.

Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND). Usage and distribution for commercial purposes as well as any distribution of modified material requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.