scholarly journals Impaired Platelet Responses to Thromboxane a2 and Thrombin in Mice Lacking Receptor-Interacting Protein Kinase 3

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2762-2762
Author(s):  
Yiwen Zhang ◽  
Jian Zhang ◽  
Rong Yan ◽  
Jie Zhang ◽  
Mengxing Chen ◽  
...  

Abstract Objective: Receptor-interacting protein 3 (RIP3) is a member of RIP family with a Ser/Thr protein kinase domain in its amino-terminus which is essential for kinase activity and autophosphorylation. The roles of RIP3 in embryonic development and different disease pathologies, such as inflammation and infections, have been reported in recent years. However, the role of RIP3 in thrombosis and hemostasis remains unknown. Methods: Hematologic analysis was performed and tail bleeding time was monitored. Mouse platelets were isolated from anti-coagulated whole blood. Platelet aggregation and secretion were recorded at real time. Platelet P-selectin exposure and specific fibrinogen binding were detected by flow cytometry. TXA2 generation was measured with enzyme immunoassay (EIA) kit. Protein phosphorylations were detected by western blotting. Result: RIP3-/- mice had tail-bleeding times that were significantly prolonged compared with their wild type littermates. In an in vivo model of mesenteric arteriole thrombosis, mice lacking RIP3 exhibited delayed thrombus formation, fewer accumulated platelets, smaller thrombi, and prolonged occlusion times. RIP3 was expressed in both human and mouse platelets. Deletion of RIP3 in mouse platelets caused a marked defect in aggregation and attenuated dense granule secretion in response to low doses of thrombin or a thromboxane A2 (TXA2) analogue, U46619. The defect in ADP secretion appears responsible for the impaired platelet aggregation, because addition of exogenous ADP rescued the reduced platelet aggregation. Although TXA2 generation and α-granule secretion were not impaired, integrin αIIbβ3 activation was attenuated in RIP3-/- platelets. Moreover, phosphorylation of Akt induced by U46619 or thrombin was markedly reduced in the absence of RIP3. Activation of Akt signaling restored the impaired aggregation of RIP3-/- platelets. ERK and p38 phosphorylation elicited by either U46619 or thrombin was attenuated in RIP3-/- platelets. In contrast, U46619- and thrombin-induced activation of PTEN, PDK1, or Src was not impaired in RIP3-/- platelets. Conclusion: Our data demonstrate a novel role for RIP3 in amplifying U46619- and thrombin-induced platelet activation by mediating Akt-dependent ADP secretion, and in supporting hemostasis and thrombus formation in vivo. RIP3 may represent a novel target to modulate PARs and TP signaling and a potential new target for antithrombotic strategy. Disclosures No relevant conflicts of interest to declare.

2017 ◽  
Vol 114 (11) ◽  
pp. 2964-2969 ◽  
Author(s):  
Yiwen Zhang ◽  
Jian Zhang ◽  
Rong Yan ◽  
Jingluan Tian ◽  
Yang Zhang ◽  
...  

Previous studies have shown that receptor-interacting protein kinase 3 (RIP3) is involved in many important biological processes, including necroptosis, apoptosis, and inflammation. Here we show that RIP3 plays a critical role in regulating platelet functions and in vivo thrombosis and hemostasis. Tail bleeding times were significantly longer in RIP3-knockout (RIP3−/−) mice compared with their wild-type (WT) littermates. In an in vivo model of arteriole thrombosis, mice lacking RIP3 exhibited prolonged occlusion times. WT mice repopulated with RIP3−/− bone marrow-derived cells had longer occlusion times than RIP3−/− mice repopulated with WT bone marrow-derived cells, suggesting a role for RIP3-deficient platelets in arterial thrombosis. Consistent with these findings, we observed that RIP3 was expressed in both human and mice platelets. Deletion of RIP3 in mouse platelets caused a marked defect in aggregation and attenuated dense granule secretion in response to low doses of thrombin or a thromboxane A2 analog, U46619. Phosphorylation of Akt induced by U46619 or thrombin was diminished in RIP3−/− platelets. Moreover, RIP3 interacted with Gα13. Platelet spreading on fibrinogen and clot retraction were impaired in the absence of RIP3. RIP3 inhibitor dose-dependently inhibited platelet aggregation in vitro and prevented arterial thrombus formation in vivo. These data demonstrate a role for RIP3 in promoting in vivo thrombosis and hemostasis by amplifying platelet activation. RIP3 may represent a novel promising therapeutic target for thrombotic diseases.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2533-2533
Author(s):  
Akruti Patel ◽  
Soochong Kim ◽  
John Kostyak ◽  
Rachit Badolia ◽  
Carol Dangelmaier ◽  
...  

Abstract PI3-kinase (phosphoinositide 3-kinase) is an important signaling molecule that is activated downstream of various receptors upon platelet activation. PI3-kinase activation leads to the generation of PIP3 (Phosphatidylinositol (3,4,5)-trisphosphate) subsequently leading to the recruitment of PH (pleckstrin homology) domain containing proteins to the plasma membrane. Our laboratory screened for proteins that interacted with PIP3 (Phosphatidylinositol (3,4,5)-trisphosphate) using PIP3 beads. One of the proteins that interacted with PIP3 was ELMO1 (Engulfment and cell motility-1). ELMO1 is a scaffold protein with no catalytic activity and is well known to regulate actin cytoskeletal rearrangement via Rac1 in other cells. However, it is not known whether ELMO1 is expressed in platelets and if so, does it regulate platelet functional responses. Here, we show that ELMO1 is present in both human and murine platelets. We used ELMO1-deficient (ELMO1-/-) mice to study its role in platelets. ELMO1-/- murine platelets showed enhanced platelet aggregation and dense granule secretion in response to the GPVI agonist, CRP (Figure 1 A & B), compared to the wildtype controls although there was no difference in GPVI expression levels between the two. There was no difference observed in response to AYPGKF- or 2-MeSADP. These data suggest that ELMO1 plays a specific role downstream of GPVI pathway but GPCRs. Moreover, ELMO1-/- platelets exhibited enhanced clot retraction and spreading indicating its role in Glycoprotein IIb/IIa (GPIIb/IIIa) mediated outside-in signaling. Furthermore, whole blood from ELMO1-/- mice perfused over collagen under arterial shear conditions exhibited enhanced thrombus formation. In an in vivo pulmonary thromboembolism model, ELMO1-/- mice showed reduced survival compared to the wildtype control. ELMO1-/- mice also showed shorter time to occlusion and increased thrombus stability using the ferric-chloride injury model indicating the role of ELMO1 in thrombus formation in vivo. At the molecular level, Rac1 activity was enhanced in ELMO1-/- murine platelets compared to the wildtype control in response to CRP (Figure 1C). Together, these data suggest that ELMO1 regulates Rac1 activity upon GPVI-mediated thrombus formation and it may play a negative regulator role in both inside-out and outside-in signaling, which might involve Rac1. Figure 1 Representative figure of (A) platelet aggregation and (B) dense granule secretion. (C) Washed platelets were stimulated with CRP 1.25 μg/mL for the indicated times. GST-PAK-RBD was used to pull-down active Rac1 from platelet lysates and was detected using specific antibody to Rac1 by Western blot. WT = Wildtype mice. ELMO1-/- = ELMO1-deficient mice. CRP = collagen related protein. Figure 1. Representative figure of (A) platelet aggregation and (B) dense granule secretion. (C) Washed platelets were stimulated with CRP 1.25 μg/mL for the indicated times. GST-PAK-RBD was used to pull-down active Rac1 from platelet lysates and was detected using specific antibody to Rac1 by Western blot. WT = Wildtype mice. ELMO1-/- = ELMO1-deficient mice. CRP = collagen related protein. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 10 (20) ◽  
pp. 4743
Author(s):  
Preeti Kumari Chaudhary ◽  
Sanggu Kim ◽  
Soochong Kim

Arrestins in concert with GPCR kinases (GRKs) function in G protein-coupled receptor (GPCR) desensitization in various cells. Therefore, we characterized the functional differences of arrestin3 versus arrestin2 in the regulation of GPCR signaling and its desensitization in platelets using mice lacking arrestin3 and arrestin2. In contrast to arrestin2, platelet aggregation and dense granule secretion induced by 2-MeSADP, U46619, thrombin, and AYPGKF were significantly potentiated in arrestin3-deficient platelets compared to wild-type (WT) platelets, while non-GPCR agonist CRP-induced platelet aggregation and secretion were not affected. Surprisingly, in contrast to GRK6, platelet aggregation induced by the co-stimulation of serotonin and epinephrine was significantly potentiated in arrestin3-deficient platelets, suggesting the central role of arrestin3 in general GPCR desensitization in platelets. In addition, the second challenge of ADP and AYPGKF restored platelet aggregation in arrestin3-deficient platelets but failed to do so in WT and arrestin2-deficient platelets, confirming that arrestin3 contributes to GPCR desensitization. Furthermore, ADP- and AYPGKF-induced Akt and ERK phosphorylation were significantly increased in arrestin3-deficient platelets. Finally, we found that arrestin3 is critical for thrombus formation in vivo. In conclusion, arrestin3, not arrestin2, plays a central role in the regulation of platelet functional responses and thrombus formation through general GPCR desensitization in platelets.


Blood ◽  
2011 ◽  
Vol 118 (2) ◽  
pp. 416-424 ◽  
Author(s):  
Olga Konopatskaya ◽  
Sharon A. Matthews ◽  
Matthew T. Harper ◽  
Karen Gilio ◽  
Judith M. E. M. Cosemans ◽  
...  

Abstract Platelets are highly specialized blood cells critically involved in hemostasis and thrombosis. Members of the protein kinase C (PKC) family have established roles in regulating platelet function and thrombosis, but the molecular mechanisms are not clearly understood. In particular, the conventional PKC isoform, PKCα, is a major regulator of platelet granule secretion, but the molecular pathway from PKCα to secretion is not defined. Protein kinase D (PKD) is a family of 3 kinases activated by PKC, which may represent a step in the PKC signaling pathway to secretion. In the present study, we show that PKD2 is the sole PKD member regulated downstream of PKC in platelets, and that the conventional, but not novel, PKC isoforms provide the upstream signal. Platelets from a gene knock-in mouse in which 2 key phosphorylation sites in PKD2 have been mutated (Ser707Ala/Ser711Ala) show a significant reduction in agonist-induced dense granule secretion, but not in α-granule secretion. This deficiency in dense granule release was responsible for a reduced platelet aggregation and a marked reduction in thrombus formation. Our results show that in the molecular pathway to secretion, PKD2 is a key component of the PKC-mediated pathway to platelet activation and thrombus formation through its selective regulation of dense granule secretion.


2014 ◽  
Vol 2014 ◽  
pp. 1-11 ◽  
Author(s):  
Dong-Seon Kim ◽  
Hyun Dong Ji ◽  
Man Hee Rhee ◽  
Yoon-Young Sung ◽  
Won-Kyung Yang ◽  
...  

Ethnopharmacological Relevance.Morus albaL. leaves (MAE) have been used in fork medicine for the treatment of beriberi, edema, diabetes, hypertension, and atherosclerosis. However, underlying mechanism of MAE on cardiovascular protection remains to be elucidated. Therefore, we investigated whether MAE affect platelet aggregation and thrombosis.Materials and Methods. The anti-platelet activity of MAE was studied using rat platelets. The extent of anti-platelet activity of MAE was assayed in collagen-induced platelet aggregation. ATP and serotonin release was carried out. The activation of integrinαIIbβ3and phosphorylation of signaling molecules, including MAPK and Akt, were investigated with cytofluorometer and immunoblotting, respectively. The thrombus formationin vivowas also evaluated in arteriovenous shunt model of rats.Results. HPLC chromatographic analysis revealed that MAE contained rutin and isoquercetin. MAE dose-dependently inhibited collagen-induced platelet aggregation. MAE also attenuated serotonin secretion and thromboxane A2formation. In addition, the extractin vivoactivity showed that MAE at 100, 200, and 400 mg/kg significantly and dose-dependently attenuated thrombus formation in rat arterio-venous shunt model by 52.3% (P<0.001), 28.3% (P<0.01), and 19.1% (P<0.05), respectively.Conclusions. MAE inhibit platelet activation, TXB2 formation, serotonin secretion, aggregation, and thrombus formation. The plant extract could be considered as a candidate to anti-platelet and antithrombotic agent.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2168-2168
Author(s):  
Lu Wang ◽  
Yi Wu ◽  
Junsong Zhou ◽  
Syed S. Ahmad ◽  
Bulent Mutus ◽  
...  

Abstract Abstract 2168 Several members of the protein disulfide isomerase family of enzymes are important in platelet function and in thrombosis. Platelet protein disulfide isomerase (PDI) has been shown to have an important role in platelet function but is reported to not be required for thrombus formation in vivo. A novel platelet PDI called ERp57 mediates platelet aggregation but its role in thrombus formation is unknown. To determine the specific role of platelet-derived ERp57 in hemostasis and thrombosis we generated a megakaryocyte/platelet specific knockout. Despite normal platelet counts and platelet glycoprotein expression, mice with ERp57-deficient platelets had prolonged tail-bleeding times and thrombus occlusion times, and defective activation of the αIIbβ3 integrin and platelet aggregation. The aggregation defect was corrected by addition of exogenous ERp57 implicating surface ERp57 in platelet aggregation. Platelet surface ERp57 protein and activity increased substantially with platelet activation. We conclude that platelet-derived ERp57 is required for hemostasis and thrombosis and platelet function. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1060-1060
Author(s):  
Soochong Kim ◽  
Carol Dangelmaier ◽  
Dheeraj Bhavanasi ◽  
Shu Meng ◽  
Hong Wang ◽  
...  

Abstract We investigated the mechanism of activation and functional role of a hitherto uncharacterized signaling molecule, RhoG, in platelets. RhoG is a ubiquitously expressed member of the Rho Family of GTPases. We demonstrated for the first time the expression [Fig 1A] and activation of RhoG [Fig 1B] in platelets. Platelet aggregation and dense-granule secretion in response to glycoprotein VI (GPVI) agonists, collagen-related peptide (CRP) and convulxin were significantly inhibited in RhoG-deficient platelets compared to wild type murine platelets [Fig 1C]. Integrin αIIbβ3 activation and α-granule secretion as measured by flow cytometry were also significantly inhibited in RhoG-deficient murine platelets downstream of GPVI agonists. In contrast, 2-MeSADP- and AYPGKF-induced platelet aggregation and secretion [Fig 1D] were minimally affected in RhoG deficient platelets, indicating that the function of RhoG in platelets is GPVI-specific.Figure 1(A): Increasing amounts of human platelet lysate (in μg) were separated by SDS-PAGE, Western blotted, and probed with anti-RhoG antibody. (B) RhoG activation was measured upon stimulation of washed human platelets with 5μg/ml CRP for various times. Washed platelets were lysed and active GTP-bound RhoG was determined by pull-down analysis using bacterially expressed GST-ELMO. (C) Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with GPVI agonists, 2.5 μg/ml CRP and 100 ng/ml convulxin and (D) G protein coupled receptor agonists, 30 nM 2MeSADP and 100 μM AYPGKF for 3.5 min under stirring conditions. Platelet aggregation and ATP secretion were measured by aggregometry.Figure 1. (A): Increasing amounts of human platelet lysate (in μg) were separated by SDS-PAGE, Western blotted, and probed with anti-RhoG antibody. (B) RhoG activation was measured upon stimulation of washed human platelets with 5μg/ml CRP for various times. Washed platelets were lysed and active GTP-bound RhoG was determined by pull-down analysis using bacterially expressed GST-ELMO. (C) Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with GPVI agonists, 2.5 μg/ml CRP and 100 ng/ml convulxin and (D) G protein coupled receptor agonists, 30 nM 2MeSADP and 100 μM AYPGKF for 3.5 min under stirring conditions. Platelet aggregation and ATP secretion were measured by aggregometry. CRP-induced phosphorylations of Syk, Akt and ERK, but not Src family kinases (SFKs), were significantly reduced in RhoG-deficient platelets compared to those of wild type [Fig 2A]. Consistently, CRP-induced RhoG activation was abolished by pan-SFK inhibitor but not by Syk or PI 3-kinase inhibitors [Fig 2B]. Interestingly, unlike CRP, platelet aggregation and Syk phosphorylation induced by fucoidan, a CLEC-2 agonist, were unaffected in RhoG deficient platelets [Fig 2C].Figure 2(A): Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with 2.5 μg/ml CRP and at 37 °C for 2 min and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Src (Tyr416), anti-phospho-Akt (Ser473), anti-phospho-ERK, or anti-β-actin (lane loading control) antibodies by western blotting. (B): RhoG activation induced by 5μg/ml CRP for 60 sec was evaluated in the presence and absence of 10 μM PP2, 2 μM OXSI-2, or 100nM wortmannin. (C): Wild type and RhoG-deficient platelets were stimulated with 100 μg/ml fucoidan and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Akt (Ser473), or anti-β-actin (lane loading control) antibodies by western blotting.Figure 2. (A): Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with 2.5 μg/ml CRP and at 37 °C for 2 min and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Src (Tyr416), anti-phospho-Akt (Ser473), anti-phospho-ERK, or anti-β-actin (lane loading control) antibodies by western blotting. (B): RhoG activation induced by 5μg/ml CRP for 60 sec was evaluated in the presence and absence of 10 μM PP2, 2 μM OXSI-2, or 100nM wortmannin. (C): Wild type and RhoG-deficient platelets were stimulated with 100 μg/ml fucoidan and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Akt (Ser473), or anti-β-actin (lane loading control) antibodies by western blotting. Finally, RhoG -/- mice had a significant delay in time to thrombotic occlusion in cremaster arterioles compared to wild type littermates [Fig 3A and 3B], indicating the important in vivo functional role of RhoG in platelets.Figure 3(A): Time required for occlusion of cremaster arterioles in RhoG +/+ and RhoG -/- mice was measured using microvascular thrombosis model with light/dye-induced injury. 5 mice of each genotype were used, and statistical analysis revealed a significant difference between the 2 genotypes of mice (*, P < .01). (B) Representative images of cremaster arterioles were taken from RhoG +/+ and RhoG -/- mice 30 min after the injury. As seen with the outline (arrows) of the thrombus formed, thrombus formation was inhibited in RhoG -/- mice.Figure 3. (A): Time required for occlusion of cremaster arterioles in RhoG +/+ and RhoG -/- mice was measured using microvascular thrombosis model with light/dye-induced injury. 5 mice of each genotype were used, and statistical analysis revealed a significant difference between the 2 genotypes of mice (*, P < .01). (B) Representative images of cremaster arterioles were taken from RhoG +/+ and RhoG -/- mice 30 min after the injury. As seen with the outline (arrows) of the thrombus formed, thrombus formation was inhibited in RhoG -/- mice. In conclusion, we show for the first time that RhoG is expressed and activated in platelets, plays an important role in GPVI/FcRγ-mediated platelet activation and is critical for thrombus formation in vivo. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 115 (20) ◽  
pp. 4083-4092 ◽  
Author(s):  
Frédéric Adam ◽  
Alexandre Kauskot ◽  
Paquita Nurden ◽  
Eric Sulpice ◽  
Marc F. Hoylaerts ◽  
...  

Abstract The role of c-Jun NH2-terminal kinase 1 (JNK1) in hemostasis and thrombosis remains unclear. We show here, with JNK1-deficient (JNK1−/−) mice, that JNK1 plays an important role in platelet biology and thrombus formation. In tail-bleeding assays, JNK1−/− mice exhibited longer bleeding times than wild-type mice (396 ± 39 seconds vs 245 ± 32 seconds). We also carried out in vitro whole-blood perfusion assays on a collagen matrix under arterial shear conditions. Thrombus formation was significantly reduced for JNK1−/− platelets (51%). In an in vivo model of thrombosis induced by photochemical injury to cecum vessels, occlusion times were 4.3 times longer in JNK1−/− arterioles than in wild-type arterioles. Moreover, in vitro studies carried out in platelet aggregation conditions demonstrated that, at low doses of agonists, platelet secretion was impaired in JNK1−/− platelets, leading to altered integrin αIIbβ3 activation and reduced platelet aggregation, via a mechanism involving protein kinase C. JNK1 thus appears to be essential for platelet secretion in vitro, consistent with its role in thrombus growth in vivo. Finally, we showed that ERK2 and another isoform of JNK affect platelet aggregation through 2 pathways, one dependent and another independent of JNK1.


Blood ◽  
2016 ◽  
Vol 127 (7) ◽  
pp. 927-937 ◽  
Author(s):  
Ming Yue ◽  
Dongjiao Luo ◽  
Shanshan Yu ◽  
Pu Liu ◽  
Qi Zhou ◽  
...  

Key Points MINK1 promotes hemostasis and thrombosis in vivo. MINK1 specifically regulates platelet dense-granule secretion.


Sign in / Sign up

Export Citation Format

Share Document