Platelet FcγRIIA Signaling Results in Ubiquitination and Cellular Translocation of Activated Syk

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2761-2761
Author(s):  
Shaji Abraham ◽  
Leonard C. Edelstein ◽  
Chad A Shaw ◽  
Pierrette Andre ◽  
Xianguo Kong ◽  
...  

Abstract Platelet FcγRIIA is central to the pathophysiology of immune-mediated thrombocytopenia and thrombosis syndromes, such as heparin-induced thrombocytopenia (HIT). FcγRIIA is also the major transmembrane signaling adapter for αIIbβ3 outside-in signaling. In HIT, antibody to heparin/PF4 is necessary but not sufficient for disease to occur. Inter-individual variation in platelet activation via FcγRIIA contributes to HIT risk, but the molecular basis for the variation is incompletely understood. In our PRAX1 study of platelet reactivity and RNA expression (Edelstein, Nature Med 2013; Simon, Blood 2014), we identified differentially expressed mRNAs from healthy donors with different platelet reactivity to FcγRIIA stimulation. We observed significant differential expression of molecules involved in ubiquitination processes in relation to platelet reactivity to FcγRIIA stimulation. Syk is a protein tyrosine kinase and the major signaling node downstream of platelet receptors that use immunotyrosine activation motif (ITAM) signaling, such as FcγRIIA, GPVI and CLEC-2. We previously reported Syk ubiquitination following GPVI stimulation, and the role of c-cbl as the E3 ubiquitin ligase (Dangelmaier, Blood 2005). Ubiquitination is an important post-translational modification that modulates signal transduction by regulating the activity, subcellular localization or stability of proteins. We tested the hypothesis that ubiquitination participates in signaling, and examined ubiquitination of Syk downstream of platelet FcγRIIA activation. Using both washed human platelets and HEL cells, we observed ubiquitination of Syk upon FcγRIIA engagement by cross-linking IV.3 mAb (10 ug/ml) with goat anti-mouse Fab’2 (30 ug/ml). Both tyrosine phosphorylation and ubiquitination of Syk occurred within 15 sec, peaked by 1-3 min and decreased thereafter. The pattern of ubiquitination was consistent with 1 to 3 Ub molecules per Syk molecule. Ubiquitinated-Syk (Ub-Syk) was increased in the presence of PR-619, a deubiquitinating enzyme inhibitor, confirming ubiquitination of Syk. Ub-Syk associates with the cytoskeletal-rich platelet fraction, membrane skeleton fraction and with cytosolic fraction in detergent lysed platelets that were fractionated by lower g-forces (15,500 x g) and higher g-forces (100,000 x g). This suggests that Ub-Syk is translocated into all cellular compartments upon platelet activation. Ub-Syk was absent upon pre-treatment with Src-family kinase inhibitor PP2 (10 uM), but minimally affected in the presence of Syk inhibitor PRT318 (1 uM), in both platelets and HEL cells, as compared to DMSO treated control cells. Further, phosphorylation of c-cbl was inhibited strongly by PP2, but only slightly inhibited by PRT318, suggesting that ubiquitination of Syk depends on Src kinase activity. Of note, Ub-Syk was not degraded by the proteasome, since no accumulation of Ub-Syk was observed by pretreatment with proteasome inhibitors MG132 or Epoxomicin in either platelets or HEL cells, compared to control cells. In conclusion, Syk is ubiquitinated upon cross-linking platelet FcγRIIA and is translocated to all major subcellular compartments. Since ubiquitinated Syk is activation-dependent and not subject to proteasomal degradation, it likely serves as a novel adapter molecule for protein-protein interactions in mediating platelet activation via FcγRIIA. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 575-575
Author(s):  
Daria Madeeva ◽  
Valerie Tutwiler ◽  
Douglas B. Cines ◽  
Mortimer Poncz ◽  
Lubica Rauova

Abstract Thrombosis is the most striking complication of heparin-induced thrombocytopenia (HIT). We have shown that monocytes are preferentially targeted by HIT antibodies because of the higher affinity of monocyte surface glycosaminoglycans (GAGs) for PF4 than the chondroitin sulfate GAGs on platelets. The contribution and mechanism by which monocytes promote thrombosis in HIT have not been fully elucidated. It has been reported that HIT antibodies activate monocytes through FcγRI and then by the MEK1-ERK1/2 intracellular pathway leading to the expression of tissue factor (TF) (Kasthuri et al., Blood 2012 119:5285). While activation of platelets by HIT antibodies through FcγRIIA is well established, the role of this receptor in monocyte activation in HIT is not clear. We examined the role of monocytes and their family of Fcγ receptors in HIT using several in vitro models, including a novel microfluidic system that allowed us to examine the prothrombotic pathways using human- and murine-based systems. Our studies showed that monocytes were key to the prothrombotic state; simply adding monocytes coated with PF4 and pre-activated with KKO, a HIT-like monoclonal antibody, was sufficient to form platelet-fibrin clots in reconstituted blood samples combining isolated red cells, platelets and mononuclear cells. Using three separate approaches, we also found that HIT antibodies bound to surface PF4/GAG complexes activate monocytes via the same Fc receptor that mediates platelet activation, i.e., FcγRIIA. First, using a strategy of blocking individual classes of Fcγ receptors known to be present on monocytes - FcγRI, FcγRIIA and FcγRIII - by monoclonal antibodies, we found that only anti-FcγRIIA decreased fibrin deposition (by 52 ± 8%; p<0.005 compared to non-blocked control). Second, activation of human platelets added to platelet depleted “whole blood” containing transgenic murine monocytes expressing human FcγRIIA was markedly higher than activation by monocytes lacking FcγRIIA, as measured by P selectin expression (2 ± 0.2 times higher) and annexin V binding (3 ± 2 times higher). Third, blocking the signaling pathway downstream of FcγRIIA selectively in monocytes by the Syk-specific tyrosine kinase inhibitor PRT318 abrogated the prothrombotic effect of monocytes as demonstrated by suppression of fibrin formation. These data add to our understanding of how monocyte activation promotes thrombosis in HIT. According our current model of platelet transactivation by monocytes, HIT immune complexes engage FcγRIIA both on platelets and on monocytes, leading to the activation of a common Syk-dependent pathway. In monocytes this leads to TF expression and thrombin generation. Thrombin generated by monocytes activates G protein-coupled receptors on platelets, while surface-bound HIT immune complexes activate platelets directly through FcγRIIA coupled to the immunoreceptor tyrosine-based activation motif pathway. Concurrent platelet activation via these two pathways is known to result in highly reactive COATED platelets. We believe the formation of a large population of COATED platelets contributes to the intensely prothrombotic nature of HIT. These studies highlight the importance of blocking FcγRIIA and its downstream signal pathways in monocytes as well as in platelets in order to develop rational strategies to attenuate the risk of thrombosis in patients with HIT. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1436-1436
Author(s):  
Jennifer Yeung ◽  
Pilar Fernandez-Perez ◽  
Joanne Vesci ◽  
Theodore R Holman ◽  
Jin Ren ◽  
...  

Abstract Platelet-mediated thrombosis is the primary underlying mechanism leading to cardiovascular life-threatening clinical events. Control of excessive platelet responses is an essential aspect of antithrombotic therapy. A number of anti-platelet drugs have been developed to target specific signaling pathways or endpoints involved in platelet activation. Despite the effectiveness of current anti-platelet therapies, uncontrolled thrombosis or bleeding complications still persist. We had proposed a potential novel therapeutic approach by which oxylipins generated by 12-lipoxygenase (12-LOX) oxidation of ω-6 could modulate platelet reactivity. We observed 12-hydroxyeicosatrienoic acid (12-HETrE), a 12-LOX oxidized oxylipin of ω-6 polyunsaturated fatty acid, dihomo-γ-linolenic acid (DGLA), significantly attenuated human platelet activation. We then verified that DGLA oxidation to 12-HETrE depended on functional platelet 12-lipoxygenase (12-LOX) in our transgenic mouse model deficient in 12-LOX enzyme in the platelets (12-LOX-/-). To determine whether 12-HETrE could be inducing its inhibitory regulation in a GPCR-like manner by which it could potentially be behaving similarly to prostacyclin to activate adenylyl cyclase and increase cAMP through the Gs pathway, we measured cAMP level in the presence of 12-HETrE. We observed 12-HETrE significantly increased cAMP levels. We investigated a downstream effector of cAMP, such as VASP 157 phopshorylation, which is a PKA substrate. Also observed both Rap1 and GPIIbIIa activation to be attenuated in the presence 12-HETrE, confirming our aggregation result. This the first study to show the signaling mechanism of 12-HETrE which is dependent on active 12-LOX oxidation of DGLA to regulate platelet reactivity in a Gs-like manner. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 411-411 ◽  
Author(s):  
Yuhang Zhou ◽  
Shaji Abraham ◽  
Leonard C. Edelstein ◽  
Carol Dangelmaier ◽  
Alexander Tsygankov ◽  
...  

Abstract Heparin-induced thrombocytopenia (HIT) is a life-threatening disease in which IgG antibodies against the heparin-PF4 complex activate platelets via FcγRIIA. We previously reported that TULA-2 serves as a negative regulator of FcγRIIA pathway by dephosphorylating Syk in HEL cells. To further investigate the effect of TULA-2 on the FcγRIIA pathway and HIT, we crossed TULA-2-/- with FcγRIIA+/+ mice. Ablation of TULA-2 resulted in hyperphosphorylation of Syk, LAT, and PLCγ2 in platelets after FcγRIIA activation. Integrin activation, calcium mobilization, and P-selectin exposure were also enhanced in TULA-2-/- murine platelets compared to TULA-2+/+. Further, anti-GPIX antibody-induced HIT-like thrombocytopenia and thrombin generation were also augmented in TULA-2-/- mice (Figure A). We also found that decreased TULA-2 level shortened tail-bleeding time in mice (Figure B), suggesting a role of TULA-2 in physiological hemostasis. Additionally, TULA-2 KO and WT platelets did not show significant differences in platelet spreading and clot retraction, indicating that outside-in signaling is not affected by the absence of TULA-2. At the protein level, TULA-2 heterozygous knockout (TULA-2+/-) platelets express 50% as much protein as their wildtype counterparts. Interestingly, TULA-2+/- mice showed significantly increased platelet reactivity and more severe thrombocytopenia in vivo compared with TULA-2+/+ mice. Together the data indicate that not only the absence of TULA-2, but also the relative level of TULA-2 expression modulate FcγRIIA-mediated platelet reactivity, HIT pathogenesis, and hemostasis. Considering that TULA-2 is also a key regulator in platelet GPVI pathway, measuring TULA-2 expression may be a valuable predictor for HIT susceptibility and platelet reactivity in general. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1066-1066
Author(s):  
Liping Ma ◽  
Luo Xianming ◽  
Hongyu Li ◽  
Yiqing Li ◽  
Xie Shuangfen ◽  
...  

Abstract Abstract 1066 Objective: Group B Streptococcus (GBS), or Streptococcus agalactiae, is one of the most common cause of life-threatening sepsis in human newborns and immunosuppressed population. Patients suffering from sepsis often display low platelet counts characterized by thrombocytopenia as a result of platelet activation. Platelets, best known for their maintenance of hemostasis, have retained features of archetypal inflammatory cells corresponding to multiple roles in antimicrobial host defense. Nine members of the Toll-like receptors (TLRs) have been reported in human platelets, and TLRs in platelets play a fundamental role in both the initiation and propagation of the inflammatory response to microbial infection. We reported previously that LPS induces platelets activation and enhances platelets to release tumor necrosis factor-α by TLR 4. Since few confirm that platelets were activated by TLRs in directly binding to bacteria, the mechanism of activation of platelets in the interaction with pathogens remains unclear. Our research aimed to verify the hypothesis that the platelets selectively bind or adhere strains of GBS and aggregate, and to investigate the mechanism through which platelets become activated in sepsis upon binding to GBS and platelet TLR 2 involves in the process. Methods: Six strains of GBS were separated from septic patients. We determined GBS inducible platelet reactivity by using light transmission aggregometry, platelet bacterial adhesion assay, FCM and scanning electron microscopy. TLR2 expression was determined by RT-PCR and western blot assay. Results: After incubating platelets with strains of GBS separately, only three strains of GBS could induce platelets activation (platelets aggregation, adhesion and upgrated the expression of CD62P on platelet, Figure 1), meanwhile the activating ability and lag time were strain-dependent. Strains of GBS that were able to induce platelets activation also upregulated the expression of platelets TLR2 (in levels of RT-PCR and protein, Figure 2). However incubated with TLR 2 antibody, platelets no longer aggregated by the stimulation of GBS (Figure 3), suggesting TLR2 was involved in GBS induced platelets activation Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3729-3729
Author(s):  
John C Kostyak ◽  
Benjamin R Mauri ◽  
Carol A Dangelmaier ◽  
Akruti Patel ◽  
Monica Wright ◽  
...  

Abstract Platelet activation is essential for hemostasis. Central to platelet activation are the signals transmitted through surface receptors like GPVI, the protease activated receptors (PARs), and C-type lectin-like receptor 2 (CLEC-2). GPVI is an ITAM-bearing receptor, while Clec-2 is a HemITAM-bearing receptor. Both ITAM and hemITAM-bearing receptors signal through spleen tyrosine kinase (Syk). Syk activity is dependent on phosphorylation of several residues including Y342, Y346, and Y519/520 (Y348, Y352, and Y525/526 in human). However, the importance of each residue on either phosphorylation of other residues or activation of Syk is unknown. Therefore, we produced a Syk Y342F knock-in mouse line via CRISPR/Cas9 to investigate the importance of Y342 on Syk-mediated signaling using a platelet system. Syk Y342F mice are viable, have no gross abnormalities, and their blood cell counts are normal. We were unable to detect any Syk Y342 phosphorylation following stimulation of Y342F platelets with the GPVI agonist collagen-related peptide (CRP) (Figure 1). Further, Platelet reactivity is reduced in response to the GPVI agonists CRP (Figure 2) and collagen, as well as the CLEC-2 agonist rhodocytin. When using CLEC-2 antibody as an agonist no response is observed in Syk Y342F mouse platelets even though WT littermate control mouse platelets responded well (Figure 3). Signaling initiated by either GPVI or CLEC-2 is also inhibited, as Syk Y519/520 phosphorylation is impaired. Furthermore, PLCg2 and SLP-76 phosphorylation are reduced in Syk Y342F platelets compared to platelets from littermate control mice following stimulation of either GPVI or CLEC-2. Although reactivity of Y342F platelets is clearly reduced compared to WT platelets, there was no difference in occlusion time following ferric chloride injury of the carotid artery. Similarly, there was no difference in mortality rate following pulmonary thromboembolism using Syk Y342F mice compared to WT littermate control mice. Nevertheless, these data demonstrate that phosphorylation of Y342 on Syk following stimulation of either ITAM or hemITAM-bearing receptors is important for Syk activation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2165-2165
Author(s):  
Shaji Abraham ◽  
Pierrette Andre ◽  
Yuhang Zhou ◽  
Leonard C. Edelstein ◽  
Chad Shaw ◽  
...  

Abstract Abstract 2165 One of the major clinical challenges in heparin-induced thrombocytopenia (HIT) is that antibodies to the heparin/PF4 complex are necessary but not sufficient to cause thrombocytopenia and thrombosis. While contributions of macromolecular antigen structure, antibody recognition and procoagulant activation of leukocytes and endothelial cells are increasingly appreciated, the sine qua non of HIT is platelet activation via FcγRIIa. It has been recognized for over 20 years that there is substantial inter-individual variation in human platelet reactivity to IgG immune complex agonists (such as in HIT) acting via FcγRIIa. In the course of our Platelet RNA And eXpression-1 (PRAX1) study on the genomics of platelet reactivity, we observed differential reactivity among 154 healthy donors of platelets when platelet-rich plasma aggregated in response to the agonist anti-CD9, a well-established model of FcγRIIa-mediated activation relevant to HIT. Importantly, even after accounting for the known sources of variation in reactivity - level of platelet surface FcγRIIa expression and for the H/R131 ligand-binding polymorphism - most of the variation in platelet reactivity remained unexplained. In parallel, we established human FcγRIIa transgenic mouse lines of the C57Bl6 strain and the 129SvJ strain. Unexpectedly, we identified differential reactivity to FcγRIIa-mediated platelet activation between these different mouse strains. The availability of models of variation in FcγRIIa-mediated platelet activation in both humans and mice allowed us a unique opportunity to test the hypothesis that differential expression (DE) of platelet RNA species accounts for differential reactivity. Total RNA was isolated from leukocyte-depleted platelets from each human donor and from each transgenic mouse strain. Using the species-appropriate Nanostring digital quantification assay, microRNAs (miRs) expressed in platelets were identified. 4 DE miRs in humans and 7 in mice were identified with a statistically significant difference in normalized expression, after Benjamini-Hochberg correction for multiple testing. DE was validated by qRT-PCR. Of note, several DE miRs were shared by human and mouse platelets. Work is ongoing as to the identity and functions of the mRNAs targeted by these DE miRs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3397-3397
Author(s):  
Aaron J Marcus ◽  
Joan HF Drosopoulos ◽  
Kim E Olson ◽  
Ranjini Anand ◽  
Dana Leifer ◽  
...  

Abstract Abstract 3397 Background: In the US, 795,000 patients are diagnosed with stroke annually. Of the 90% of strokes that are ischemic, a substantial number are of unknown etiology. In younger adults, the proportion of such cryptogenic strokes exceeds 40%. Cryptogenic stroke patients have an increased prevalence of interatrial shunts (IAS), which can be found in as many as 60% of cases as compared to 26% for the general population. Yet, percutaneous closure of such defects has not been found to reduce the recurrence rate as compared with medical therapy. Hypercoagulable disorders have also been implicated in the pathogenesis of premature ischemic strokes, but their occurrence in cryptogenic stroke series is modest, leaving many such strokes unexplained. We hypothesized that cryptogenic stroke may in part represent a disorder of enhanced platelet reactivity. An understanding of the responsiveness of platelets from young cryptogenic stroke patients to a broad panel of agonists is required because platelet reactivity to a single agonist or a thromboxane measurement does not provide sufficient information about platelet reactivity. CD39/NTPDase1, the ecto-enzyme on endothelial cells and leukocytes, is responsible for control of adenine nucleotide levels in the local vascular microenvironment, and, therefore, blood fluidity. CD39 rapidly metabolizes ATP and ADP released from activated platelets and other cells, thereby abolishing the prothrombotic cascade of platelet activation and recruitment. Methods: We sought to determine whether platelet activation and recruitment, markers of platelet activation, and CD39 enzymatic activity are increased in younger patients with unexplained stroke. To this end, we evaluated such platelet-related measures in a subsample (n=167 subjects) participating in the THrombophilia In Cryptogenic stroKe (THICK) study, a prospective case-control study evaluating the prothrombotic determinants of unexplained stroke. Cases consisted of patients, ages 18–64, referred to Weill Cornell Medical Center for evaluation of cryptogenic stroke, whereas controls were stroke-free volunteers within the same age range recruited at our center and the surrounding area. Cryptogenic stroke was defined according to modified TOAST criteria. Results: Cases (n=99) were similar in age (median 45 years old), sex (51% women), and race-ethnic composition (8% African American) as compared with controls (n=68). They also had similar prevalences of hypertension (18%), diabetes (7%), dyslipidemia (40%), and current smoking (6%). Cases had over twice the prevalence of IAS (56%) as controls (24%). Extensive testing for humoral prothrombotic disorders revealed a potential explanation for stroke in only half of the patients. We developed a Broad Range Aggregometry System to study profiles of platelet reactivity to multiple agonists, and classified patient responsiveness based on their reactivity to these agonists. We have also been able to make a laboratory diagnosis of aspirin non-responsiveness in these patients and controls. In addition, in stroke subjects, trends to higher total ADPase activity were observed on lymphocytes (p=0.09) and PMN (p=0.09), while ATPase activities were similar (p=0.81 and 0.68, respectively), as determined by our radio-TLC nucleotidase assay. Also, the ADPase to ATPase activity ratio was greater in the stroke patients than controls (p=0.003 for lymphocytes; p=0.13 for PMN). Cryptogenic stroke patients also displayed increased levels of platelet-monocyte aggregates (p=0.034) and the platelet activation marker CD63 (p=0.048), but not CD154 (p=0.183), as compared to controls (determined by FACS analyses). Conclusions: These findings support a potential role for enhanced platelet reactivity and a low threshold for platelet responsiveness in cryptogenic stroke patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4817-4817
Author(s):  
Kalpana Rao Prakasa ◽  
Lewis Becker ◽  
Nauder Faraday ◽  
Diane Becker ◽  
Rehan Qayyum

Background Clopidogrel is an antiplatelet agent that acts through direct inhibition of ADP-dependent platelet activation. Several studies have also found an inhibitory effect of clopidogrel on inflammation. Although platelets are known to participate in inflammatory processes, the mechanism(s) through which clopidogrel might modify platelet-mediated inflammation is unknown. We examined the platelet transcriptome to gain insight into the mechanism(s) through which clopidogrel modulates inflammation. Methods Three individuals with subclinical atherosclerosis and family history of early onset coronary artery disease were enrolled. Blood was collected for RNA-seq at baseline and one-week after clopidogrel 75mg daily. RNA was extracted from leukocyte-depleted platelet-rich plasma and approximately 70 million 100bp paired-end reads per sample were obtained using the Illumina HiSeq 2500. RNA-seq analysis was performed using Bowtie/TopHat/Cufflinks software pipeline. Genes with expression ≥ 0.3 fragments per kilobase of transcript per million reads (FPKM) were considered to be expressed. We set the threshold for significant differential expression as ≥ 2-fold change in gene expression after clopidogrel therapy with a false discovery rate of 0.05. Results The study sample consisted of 2 women and 1 man. Among the 12,297 expressed genes, the most highly expressed nonubiquitous genes included PPBP, PF4, and TUBB1, confirming platelets as the source of the RNA. Comparing samples before and after clopidogrel, treatment was associated with up-regulation of 733 genes and down-regulation of 387 genes. Pathway analyses found that genes related to platelet activation, degranulation, and aggregation were up-regulated, while genes related to immune system processes, such as B- and T-cell activation and innate immune response, were down-regulated. Examples of down-regulated genes include the toll-like receptor family genes, TLR2, TLR4, TLR7, and TLR8, the chemokine receptor genes CCR2, CCR5,CCR6, and CCR7, and the interleukin receptor genes, IL4R, IL7R, and IL13RA. Conclusion One week of clopidogrel therapy up-regulates transcription of pro-thrombotic platelet genes and down regulates transcription of inflammation-related platelet genes. These data suggest that clopidogrel may exert a direct anti-inflammatory action by inhibiting the ability of platelets to participate in inflammatory processes. The relationship of variability in clopidogrel-induced modulation of these pathways to clinical outcomes is unknown. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2194-2194
Author(s):  
Elizabeth E Gardiner ◽  
Jianlin Qiao ◽  
Cheewee Tan ◽  
Jane Frances Arthur ◽  
Mohammad Al-Tamimi ◽  
...  

Abstract Abstract 2194 Metalloproteinase-mediated ectodomain shedding of platelet-specific receptors for collagen (GPVI) and von Willebrand factor (GPIbα of the GPIb-IX-V complex) is triggered by either ligand-induced platelet activation-dependent pathways, or by activation-independent pathways mediated by Factor Xa or induced by the thiol-modifying agent, N-ethylmaleimide. We recently reported that shed soluble GPVI (sGPVI) was elevated in plasma of 159 ischaemic stroke patients compared with 159 community-based controls (P=0.0168), and in 29 patients with disseminated intravascular coagulation compared with healthy donors (n=25, P=0.002), consistent with a pathophysiological role for GPVI shedding from human platelets. Our new studies now show that transient exposure of human platelets to arterial or pathological shear rates of 3000–10,000 s−1 for 1–5 min ex vivo in a cone-plate viscometer, in the absence of GPVI ligand or platelet activation, activated sheddases producing a 2- to 3-fold increase in plasma sGPVI and a corresponding loss of surface GPVI from sheared platelets. Shear-induced GPVI shedding was blocked by GM6001 or GI254023, a selective inhibitor of ADAM10. In contrast to shear-induced platelet aggregation, shedding was unaffected by inhibitors of aggregation (VWF-blocking anti-GPIbα mAb, AK2, or the αIIbβ3 antagonist, RGD peptide) or by the absence of VWF in a patient with von Willebrand's disease Type III (VWF antigen levels <1%). Further, shedding of GPVI increased for up to 10 min after cessation of a short exposure of platelets to shear even when signalling, secretion and aggregation was blocked by inhibiting intracellular kinases (PP2, piceatannol), thromboxane generation (aspirin), ADP (apyrase) and calcium flux (BAPTA). Together, the combined results provide the first evidence that receptor sheddase activity can be regulated by hydrodynamic shear stress independent of cellular activation. This may represent a novel protective mechanism for down-regulating platelet reactivity as a response to pathological shear. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3254-3254
Author(s):  
Jennifer Yeung ◽  
Kenneth Ikei ◽  
Joanne Vesci ◽  
Alex Arnouk ◽  
Theodore R Holman ◽  
...  

Abstract Abstract 3254 Platelet activation plays a pivotal role in thrombosis and hemostasis. Understanding the signaling events mediating this process is essential in preventing unwanted clot formation, which can lead to the development of heart attack, stroke, and venous thromboembolism. Fatty acids may play an important role in determining the level of platelet reactivity, however the mechanism(s) by which this occurs are not entirely clear. 12-lipoxygenase (12-LOX) has been shown to oxidize the fatty acid, arachidonic acid (AA), in order to produce the bioactive eicosanoid 12-HETE, which has recently been shown to play a role in tissue factor activation and subsequent thrombin generation in the platelet (Thomas et al., 2010, J Biol Chem; 285:6891–903). 12-HETE has also been shown to signal to cells, in part, through the G protein-coupled receptor GPR31 (Guo et al, 2011, J Biol Chem; epub). While research on 12-LOX-mediated eicosanoid regulation of platelets has primarily focused on 12-HETE, the potential for regulation of platelets by eicosanoids derived from other fatty acids has been overlooked. As the fatty acid content on the platelet membrane is extremely dynamic and fatty acid supplementation is correlated with a reduced risk for cardiovascular disease, we hypothesized that 12-LOX oxidation of another fatty acid may play a direct role in regulating platelet function. To test this hypothesis, the Ω-6 fatty acid dihomo-γ-linolenic acid (DGLA) as well as its eicosanoid derived from 12-LOX oxidation, 12-hydroxyeicosatrienoic acid (12-HETrE), were applied to washed platelets followed by stimulation with thrombin, PAR1-AP, or PAR4-AP. Platelets treated with either DGLA or 12-HETrE showed significant attenuation in platelet aggregation following stimulation with thrombin or PAR-AP. A number of biochemical intermediates were also tested in the presence of 12-HETrE or DGLA including Rap1 activation, αIIbβ3 integrin activation, α-granule secretion, and dense granule secretion. All endpoints tested were attenuated in the presence of DGLA or 12-HETrE relative to control. To confirm the regulation was unique to DGLA and its metabolite, the same endpoints were measured in the presence of AA or 12-HETE. Pre-treatment with either AA or 12-HETE did not attenuate any of the agonist-mediated platelet activation endpoints. Thus, our data supports a unique role for 12-HETE and 12-HETrE and that fatty acid regulation of platelet function may be highly dependent on the lipid content of the platelet. Shifting the ratios of fatty acids in the platelet through dietary supplementation or pharmacological intervention may be sufficient to induce a cardio-protective state, in part, through increasing 12-HETrE formation and subsequently inhibiting platelet activation. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document