FcγRIIA Mediates the Prothrombotic Role of Monocytes in HIT

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 ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 224-224 ◽  
Author(s):  
Sam Glover ◽  
Nigel S. Key ◽  
Gowthami M Arepally ◽  
Nigel Mackman ◽  
Raj S. Kasthuri

Abstract Abstract 224 Introduction: Heparin-induced thrombocytopenia (HIT) is a major cause of drug-induced thrombocytopenia and occurs in 1–5% of individuals exposed to heparin. Paradoxically, 30–50% of individuals with HIT develop thrombosis. The mechanism of thrombosis in HIT is poorly understood. We recently reported that HIT antibody complexes induce tissue factor (TF) expression in monocytes and result in the release of TF-positive microparticles (MPs). The mechanism by which HIT antibody complexes induce monocyte TF has not been established. The objective of this study is to characterize the receptors involved in HIT antibody complex mediated induction of TF expression in monocytes. As HIT antibody complex mediated activation of platelets is dependent on the FcgRIIA receptor, we evaluated the role of the FcgRII receptor in the induction of monocyte TF by HIT antibody complexes. We also evaluated the role of toll like receptor-4 (TLR4) and the platelet factor 4 (PF4) chemokine receptor CXCR3 in this process. Methods: The combination of heparin, PF4 and the murine monoclonal PF4/heparin-specific antibody KKO has been shown to cause activation of platelets and monocytes, and mimic HIT in vitro. Peripheral blood mononuclear cells (PBMCs) from healthy volunteers were pre-incubated for 30 min at 37°C with an inhibitory antibody to the FcgRII receptor (IV.3); anti-CXCR2, 3, or 4 antibodies; anti-TLR4 antibody; or mouse-IgG (mIgG) control. Following pre-incubation with antibodies for 30 minutes, heparin (1U/mL), PF4 (10μg/mL), and KKO (100μg/mL) – together referred to as the HIT antibody complex – were added. Heat-aggregated mIgG and LPS were used as positive controls for the FcgRII and TLR4 receptors, respectively. Following a 6-hour incubation, PBMCs were pelleted by centrifugation and MPs were isolated from the supernatant. The procoagulant activity (PCA) of PBMCs and MPs was measured using clotting assays performed in the presence of the anti-TF antibody HTF-1 or control antibody. TF dependent PCA was calculated by reference to a standard curve generated using relipidated recombinant TF. Results: Incubation of PBMCs with heat aggregated mIgG for 6 hours resulted in significant induction of cellular TF (345 +/− 36 pg/106 cells) which was blocked by 30 min pre-incubation with the antibody IV.3 (146 +/− 17 pg/106 cells, N=3, p<.003). However, pre-incubation with IV.3 had no significant effect on TF induction (140 +/− 5 pg/106 cells) associated with the HIT antibody complex when compared to control mIgG (110 +/− 18 pg/106 cells, N=3, p<0.11). PBMCs incubated with HIT antibody complexes in the presence of a TLR-4 antibody showed less TF activity (52 +/− 4 pg/106 cells) compared to control mIgG (80 +/− 10 pg/106 cells N=3, p<0.025). A similar, partial inhibition of TF activity was also observed in PBMCs incubated with LPS in the presence of an anti-TLR4 antibody (121 +/− 3 pg/106) compared with a control antibody (89 +/− 2 pg/106, N=3, p<.0013). Experiments with a more effective inhibitor of TLR4 are in progress. PBMCs incubated with the HIT antibody complexes in the presence of an anti-CXCR3 antibody showed less TF activity (36 +/− 7 pg/mL) compared to control mIgG (118 +/− 15 pg/106 cells, N=3, p<0.004). Antibodies against CXCR2 and CXCR4 did not have any significant effect on TF induction. Measurement of MP TF activity mirrored the results described above. Using flow cytometry and an anti-CXCR3 antibody labeled with FITC, we found that 5% (± 0.5%) of monocytes expressed CXCR3 (N=3), which is consistent with the reported literature. Conclusions: These data suggest that induction of TF in monocytes by HIT antibody complexes is not mediated by the FcgRII receptor. This is contrary to the mechanism of platelet activation by these antibody complexes, which is an FcgRIIa dependent process. We found that TLR4 plays a role in HIT antibody complex mediated induction of TF in monocytes and blocking TLR4 led to a 30% decrease in TF activity. On the other hand, CXCR3 appeared to play a more significant role with blockade of CXCR3 leading to a 70% decrease in TF activity. Further characterization of the role of these receptors in HIT antibody complex mediated induction of TF expression in monocytes is required. We speculate that the extent of CXCR3 and TLR4 expression in monocytes may influence the susceptibility to developing thrombotic complications in HIT. Disclosures: No relevant conflicts of interest to declare.


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 ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4614-4614
Author(s):  
Ekaterina Mikhaltsova ◽  
Valeri G. Savchenko ◽  
Larisa A. Kuzmina ◽  
Mikhail Drokov ◽  
Vera Vasilyeva ◽  
...  

Abstract Introduction It's generally considered that all alloimmune process such as acute graft-versus host disease (aGVHD) after allo-HSCT are mostly controlled by lymphocytes. The role of neutrophils in systemic alloimmunity after allo-HSCT is still illusive. In 1987 a distinct subset of proinflammatory, low-density granulocytes (LDGs) isolated from the peripheral blood mononuclear cell fractions of patients with system lupus erythematosus has been described. There is no LDG's in healthy donors. While the origin and role of LDGs still needs to be fully characterized, we try to describe this population in patients with hematological malignancies after allo-HSCT Patients and methods. Peripheral blood samples were collected in EDTA-tubes before allo-HSCT, on day +30,+60,+90 after allo-HSCT and at day of aGVHD from 47 patients with hematological malignancies (AML=22, ALL n=17, LPD=3, MDS =2; CML=2; 17 with active disease, 30 - in CR) after allo-HSCT (from matched unrelated donor n=34, from matched related donor n=13; MAC = 13, RIC=34). Isolation of mononuclear cells from human peripheral blood was made by standard protocol using Lympholyte®-M Cell Separation Media (Cedarlane Labs). The anti-CD66b-PE (Biolegend, USA) antibodies and FSC/SSC were used to determine LDGs cells as FSChigh \SSChigh \CD66b+. 100000 of cells were analyzed on a BD FACSCanto II (Becton Dickinson, USA). Results. Results of blood evaluation of 47 patients with hematological malignancies, whose blood was examined after allo-HSCT presented in table 1. Conclusion Despite the fact that we don't get significant differences. LDG's detection in allo-HSCT patients need further investigation. Table 1. Incidence of LDG after allo-HSCT in patients with and without aGVHD Table 1. Incidence of LDG after allo-HSCT in patients with and without aGVHD Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1185-1185
Author(s):  
Lucia Catani ◽  
Stefania Lorenzini ◽  
Rosaria Giordano ◽  
Paolo Caraceni ◽  
Maria Rosa Motta ◽  
...  

Abstract Abstract 1185 Background. The potential role of bone marrow (BM)-derived stem cells (SCs) in patients with end-stage liver disease has been addressed by our group in four studies. Main objectives were: 1) to assess stem/progenitor cell mobilization in 24 patients receiving orthotopic liver transplantation (OLT); 2) to evaluate whether G-CSF can be safely administered to patients with liver cirrhosis in order to expand and mobilize BM-derived SCs; 3) to investigate the effects of transplantation of human G-CSF-mobilized CD34+ and CD133+ SCs in mice with chronic liver injury and fibrosis; 4) to evaluate the feasibility and the safety of the purification and intrahepatic reinfusion of increasing numbers of autologous BM-derived G-CSF-mobilized CD133+ SCs in patients with end-stage liver disease. Methods. 1) Flow cytometry analysis, clonogenic assays and RT-PCR have been performed after OLT; 2) 18 patients with advanced liver disease were consecutively treated with increasing doses of G-CSF starting from 2 μg/kg/daily; 3) C57BL/6N mice received CCl4 by inhalation for thirteen weeks and were treated with Cyclosporin-A. Transplantation was performed by injection (tail vein) of 106 CD34+ or CD133+ SCs of three cirrhotic patients. After four weeks from transplantation all mice were sacrificed; 4) G-CSF at 7.5μg/Kg/b.i.d. is administered subcutaneosly (sc) from day 1 until the completion of peripheral blood stem cells (PBSC) collection. Collection of PBSC will begin on day + 4 only if the concentration of CD133+ cells is 38/mL. PB mononuclear cells obtained from mobilized standard-volume leukapheresis will be incubated with Macs colloidal superparamagnetic CD133 microbeads. CliniMacs device is used for the positive selection of CD133+ SCs under GMP conditions. At least 4 weeks after SC mobilization, collection and cryopreservation, highly purified autologous G-CSF-mobilized CD133+ cells are re-infused through the hepatic artery by transfemoral or transbranchial arteriography. CD133+ cells are administered to patients starting from 5×104/Kg patient's body weight and increased every 3 patients. The maximum infused cell dose will be 1×106/kg. G-CSF at 5μg/Kg/day is administered sc for 3 days after the reinfusion of SCs for their expansion and to induce a selective proliferative advantage of reinfused cells in vivo. Results and Discussion. 1) We demonstrated that both early subsets of the hematopoietic SC compartment (CD34+/CD90+ cells) and more mature committed progenitors (CFU-C) were mobilized into PB after OLT. We also demonstrated the release from the BM of liver-committed HSCs co-expressing epithelial markers after OLT; 2) We show that the administration of G-CSF to patients with liver cirrhosis is safe and feasible and allows the mobilization and collection of BM-derived SCs at the dose of 15 mg/kg/day. 3) We demonstrated that mice transplanted with either CD133+ or CD34+ human cells appear to have less fibrotic septa than mice without SC transplantation, suggesting the potential therapeutic role of human SCs on the recovery of liver fibrosis. 4) Up to date, three patients with end stage liver disease have been successfully mobilized with G-CSF and highly purified autologous CD133+ SCs have been re-infused. The number of collected CD133+ SCs is 0,7, 0,2 and 0.35×106/Kg, respectively. The number of the re-infused highly purified CD133+ SCs is 4.7, 5.0 and 5.4×104/Kg, respectively. No adverse events have been recorded during mobilization or intrahepatic SCs re-infusion. Updated results on current patients and future patients will be presented at the Meeting. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 486-486
Author(s):  
Guoying Zhang ◽  
Binggang Xiang ◽  
Radek C. Skoda ◽  
Susan S. Smyth ◽  
Xiaoping Du ◽  
...  

Abstract Abstract 486 The role of intracellular secondary messenger cGMP in platelet activation has been controversial, with both stimulatory and inhibitory roles reported. The platelet cGMP is believed to be predominantly synthesized by soluble guanylyl cyclase (sGC), which is activated by nitric oxide (NO). To specifically determine the role of sGC-dependent cGMP synthesis in platelet function and in vivo thrombosis and hemostasis, we produced mice harboring a “floxed” sGC beta1 allele. In the “floxed” sGC beta1 mice (sGC beta1fl/fl), the exons 7 and 8 of sGC beta1 gene and an inserted Neo cassette were flanked with three LoxP sites. Platelet-specific deletion of sGC beta1fl/fl allele was accomplished through breeding of the sGC beta1fl/fl mice with pf4-Cre recombinase transgenic mice. Immunoblotting showed the complete absence of this protein in sGC beta1fl/fl/Cre platelets. Mice lacking sGC beta1 in platelets appeared to develop normally and had normal blood counts, including platelets. Blood pressure of platelet-specific sGC deficient mice was comparable to that of wild-type littermates. Inactivating the sGC beta1 gene in platelets abolished cGMP production induced by either NO donors or platelet agonists that are known to activate endogenous NO synthesis, confirming that both the platelet agonist-induced and NO donor-induced platelet cGMP production are predominantly mediated by sGC. Platelets lacking sGC exhibit a marked defect in aggregation and secretion in response to low doses of platelet agonists, collagen and thrombin. Importantly, tail-bleeding times were significantly prolonged in the platelet-specific sGC deficient mice compared with the wild-type littermates. In a FeCl3-induced carotid artery thrombosis model, time to occlusive thrombosis was prolonged in the platelet-specific sGC deficient mice, compared to wild type littermates. Thus, the agonist-stimulated sGC activation is important in promoting platelet granule secretion and aggregation. On the other hand, NO donor SNP-induced inhibition of platelet activation was abolished in sGC-deficient platelets. However, at high concentrations (>100μM), SNP inhibited platelet activation in both wild type and sGC deficient mice, indicating that both cGMP-dependent and -independent mechanisms are involved in NO donor-induced inhibition of platelet activation. Together, our data demonstrate that sGC contributes to both agonist-induced platelet activation and NO donor-induced platelet inhibition. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1669-1669
Author(s):  
Su Chu ◽  
Ravi Bhatia

Abstract Abstract 1669 Tyrosine kinase inhibitor (TKI) treatment inhibits proliferation in CML stem/progenitor cells, but only modestly increases apoptosis. Residual leukemia stem cells remain a potential source of disease relapse in IM-treated patients. The Bcl-2 family of anti-apoptotic proteins plays a central role in the regulation of apoptosis. Several Bcl-2 inhibitors are being evaluated in preclinical and clinical studies and there is considerable interest in evaluating their ability to induce apoptosis in CML stem and progenitor cells. However these agents have considerable toxicity possibly related to lack of selectivity for individual family members. We performed a functional siRNA screen to determine the role of individual Bcl-2 family members in maintaining survival of in CML and normal CD34+ cells. CML and normal CD34+ cells were transfected with siRNAs targeting Bcl-2, Bcl-2L1, Bcl-2L2, Bcl-2L10, Mcl-1 and Bcl2A1. In this screen Mcl-1 knockdown resulted in significant reduction in viability of CML CD34+ cells, with or without co-treatment with IM (1uM). Significant reduction in normal CD34+ viability was not seen. These results were validated using different siRNA sequences to knockdown Mcl1 expression. Increased apoptosis of CML but not normal CD34+ cells was seen (23±8% for CML vs. 4.2±1.5% for normal CD34+ cells, n=3, p<0.5). CML CD34+ cell apoptosis was further enhanced by combination of Mcl-1 inhibition with IM treatment (48±15% for CML vs. 7.2±3% for CB progenitors, p<0.1). To further evaluate the role of Mcl-1 in regulating CML CD34+ cell growth, an anti-Mcl-1 shRNA construct was cloned into the pHIV7-SF-RFP lentivirus vector. Cord blood and CML CD34+ cells were transduced with Mcl-1 specific or control, non-specific shRNA expressing vectors. Western blotting demonstrated effective knockdown of Mcl-1 protein levels in Mcl-1 shRNA transduced CD34+cells (82% reduction in CML and 78% in normal CD34+ cells). CD34+ RFP+ cells were selected by flow cytometry and cultured in presence and absence of IM. A significant increase in apoptosis was seen in Mcl-1 knockdown CML CD34+ cells compared with control shRNA-transduced cells, and further increase in apoptosis was seen following IM treatment (4.7±0.5 for control shRNA-transduced cells VS 25.7±2.1 for Mcl-1 knockdown cells). Mcl-1 knockdown CML CD34+ cells generated fewer colonies in methylcellulose progenitor culture (93 colonies for control siRNA transduced cells vs. 31 colonies for Mcl-1 knockdown cells) and demonstrated reduced cell expansion following culture with growth factor (SCF; IL3; GM-CSF and G-CSF) compared with control shRNA transduced cells (383,750± 172,476 for control shRNA-transduced cells 224,250± 87,044 for Mcl-1 knockdown cells). Cell expansion was further reduced with IM treatment. Mcl-1 knockdown resulted in complete loss of erythroid colony formation. Analysis of cell differentiation by flow cytometry after culture for 4 or 7 days revealed that Mcl-1 knockdown resulted in reduced generation of both erythroid (GPA+) and myeloid (CD33+ and CD14+) cells. In contrast to the results of the initial siRNA studies, shRNA-mediated Mcl-1 knockdown also resulted in significantly increased apoptosis of normal CD34+ cells (12.6± 1.6% for control shRNA-transduced cells and 24.5± 0.9% for Mcl-1 knockdown cells) associated with reduced colony formation and reduced growth in culture (1.265e+006± 273,892 for control shRNA-transduced cells 589,000 ± 188,082 for Mcl-1 knockdown cells). We conclude that RNAi-mediated Mcl-1 knockdown inhibits CML CD34+ cell survival and proliferation and enhances apoptosis after IM treatment, but also reduces viability of normal CD34+ cells. Since Mcl-1 protein expression is subject to multiple levels of regulation, our results suggest that strategies to selectively target Mcl-1 regulatory mechanisms active in CML but not normal progenitors may be less toxic and have greater clinical utility than direct targeting of the protein. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1134-1134
Author(s):  
Kelly A O'Brien ◽  
Nissim Hay ◽  
Xiaoping Du

Abstract Abstract 1134 Ligand binding to integrins mediates cell adhesion and transmits “outside in” signals that lead to cell spreading, migration, and proliferation. In platelets, the prototype integrin aIIbb3-mediated outside-in signaling is required for platelet spreading and retraction, and greatly amplifies platelet activation. Previous studies suggest that phosphoinositide 3-Kinases (PI3K) are activated upon binding of integrin αIIbβ3 to its ligand fibrinogen, and is important in outside-in signaling leading to platelet spreading. However, the mechanism by which PI3K transmits outside-in signals has been unclear. A major known downstream effector of PI3K is the Akt (protein kinase B) family of serine/threonine kinases, including Akt1, Akt2, and Akt3. We have recently shown that platelets not only express Akt1 and Akt2 as previously reported, but also express a substantial amount of Akt3. To investigate whether Akt3 is a downstream effector mediating PI3K-dependent integrin outside-in signaling, platelets from Akt3 knockout mice were compared with wild type platelets for their spreading on fibrinogen. Platelets from Akt3−/− mice showed partially, but significantly reduced spreading on fibrinogen, indicating that Akt3 is important in integrin-mediated outside-in signaling leading to platelet spreading. Consistent with the results of Akt3 knockout, treatment of platelets with a pan Akt inhibitor also significantly inhibited spreading of human and mouse platelets on fibrinogen. Akt becomes phosphorylated upon platelet spreading on fibrinogen, which is significantly reduced in Akt3 knockout platelets, and is abolished by PI3 Kinase inhibitor, wortmannin, or Src Family Kinase (SFK) Inhibitor, PP2, suggesting that Akt activation is downstream from PI3K, and SFK during integrin outside-in signaling. Thus, our data reveals that Akt3 is an important downstream effector of PI3K-dependent integrin outside-in signaling promoting platelet spreading. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3628-3628
Author(s):  
Abraham Kornberg ◽  
Marina Izak ◽  
Yossi Cohen

Abstract Treatment of multiple myeloma (MM) with thalidomide or lenalidomide is associated with increased incidence of thrombosis in contrast to treatment with bortezomib. The mechanism of thrombosis is unknown. Monocytes generate potent tissue factor (TF), the main activator of coagulation, in response to various stimuli (endotoxin, cytokins and others) and in diseases with increased incidence of thrombosis. We investigated the capacity of monocytes from patients with MM to generate TF in relation to different modalities of treatments and activation of coagulation. Peripheral blood mononuclear cells (PBMC) were isolated on Ficoll-Hypaque centrifugation and monocytes by adhesion. TF activity was assayed by modified PT using the cells as source of TF and TF antigen by ELISA, in endotoxin stimulated (10 ug/ml) and unstimulated PBMC ( all patients) and purified monocytes (50% of patients), before and after 3-4 courses of treatment. Monocytes were identified by anti CD14 and Plasma Fragment1.2 was assayed by ELISA. No TF activity@ (X10-5 U/monocyte) TF antigen@ (X10-5 pg/monocyte) Plasma fragment 1.2@ (nM) Endotoxin - + - + Bortezomib based treat* 12  before 2.2 4.8 22 64 0.28  after 1.8 4.2 18 57 0.36 Thalidomide based treat* 4  before 2.4 5.5 26 60 0.24  after 1.8 34.8# 20 440# 2.48# Lenalidomide based treat^ 6  before 2.1 6.2 28 62 0.30  after 4.1 58.6# 16 312# 3.42# @Mean *First line treatment ^Second line treatment # P<0.05 One patient on thalidomide and 2 on lenalidomide developed thrombosis. The results show that TF was low in unstimulated monocytes from all patients before treatment. It was increased mildely in stimulated monocytes from all patients before treatment and also after treatment with bortezomib (X 2-3). TF in stimulated monocytes and also plasma fragment1.2 were increased significantly after treatment with thalidomide and lenalidomide (x 14-22 and X 10). The phenomenon of enhanced capacity of monocytes to generate potent TF after treatment with thalidomide and lenalidomide but not bortezomib, and the correlation with activation of coagulation suggest a role of monocyte TF in thrombus formation by these drugs. The mechanism of the enhanced capacity is unknown and may be attributed to the immunomodulatory effect of the drugs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 10-10
Author(s):  
Jose Perdomo ◽  
Jaa Yien New ◽  
Zohra Ahmadi ◽  
Xing-Mai Jiang ◽  
Beng H Chong

Abstract Introduction. Heparin is widely used as an anticoagulant to prevent thrombosis and to treat venous thromboembolism and myocardial infarction. A complication of heparin use is the development of heparin-induced thrombocytopenia (HIT), which is a limb- and life-threatening disorder due to associated thrombotic events. HIT arises through the formation of immune complexes between heparin, platelet factor 4 and HIT autoantibodies. These immune complexes engage with FcγRIIa receptors on platelets, leading to platelet activation and aggregation and subsequent initiation of the coagulation pathway. Current HIT treatment consists of cessation of heparin administration and substitution with parenteral anticoagulants such as argatroban and danaparoid. While these anticoagulants are generally beneficial in reducing thrombocytopenia, they are only partially effective since the risk of thrombosis continues due to the underlying FcγRIIa-mediated platelet activation. Thus, alternative anticoagulants do not reduce morbidity and mortality rates, highlighting the need for more effective HIT interventions. Methods. IV.3 is a monoclonal antibody that recognizes and blocks the FcγRIIa receptor and is used in assays to confirm the presence of HIT antibodies. We derived the VH and VL sequences of IV.3 and constructed a single-chain variable fragment (scFv) antibody in the form of VH-linker-VL. Using a complementarity determining region grafting and point mutation approach the scFv was humanized with the aim of reducing potential immunogenicity for future clinical applications. The molecule was expressed in E. coli and purified by FPLC. We reconstituted the HIT condition in a micro-fluidics device on a Vena8 Fluoro+ biochip coated with vWf using whole blood flowing at 20 dyne/cm2 at 37oC. Whole blood was stained with DiOC6 and the formation of platelet aggregates was monitored by fluorescence microscopy. Video images were acquired at 1 frame every 2 sec for 460 sec. Results. The purified scFv interacts with FcγRIIa on platelets. Platelet aggregation and serotonin release assays show that the scFv effectively prevents aggregation and activation induced by HIT immune complexes. We demonstrate that in the HIT condition reconstituted in a micro-fluidics system the scFv precludes thrombus deposition in a dose-dependent manner as determined by thrombus coverage area and mean thrombus diameter (Figure 1). Conclusions. These data provide evidence that a humanized scFv binds and neutralizes FcγRIIa on platelets. This interaction prevents HIT immune complex-induced platelet aggregation and activation in vitro and stops thrombus deposition ex vivo. This molecule, therefore, inhibits a critical initiating event in HIT and may serve as a potential treatment for this condition. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document