Mechanisms of Platelet Activation by Antithymocyte Globulin (ATG).

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3905-3905
Author(s):  
Florian Langer ◽  
Francis A. Ayuk ◽  
Ali Amirkhosravi ◽  
Brigitte Spath ◽  
Todd Meyer ◽  
...  

Abstract We have previously shown that polyclonal rabbit ATG from Fresenius (ATG-F) can induce non-overt DIC in patients undergoing hematopoietic stem cell transplantation (HSCT) [Weber et al. 2003]. The compensated state of the coagulopathy suggested that the severe drop in platelet counts (>60%) observed in patients with normal pretreatment levels was not solely due to thrombin generation and platelet consumption. We thus further investigated the mechanisms of ATG-F-induced thrombocytopenia, hypothesizing that ATG-F had direct platelet-stimulating activity. Using an indirect flow cytometric binding assay we found that, compared to control IgG, ATG-F dose-dependently (1–100 μg/ml) bound to the surface of resting and TRAP-activated platelets, showing an up to 50fold increase in MFI at 50–100 μg/ml. In a washed platelet system, in which baseline positivity for CD62P was generally <15%, ATG-F alone had no effect on platelet aggregation. However, when platelets were primed with low concentrations of ADP (1–2 μM) or epinephrine (0.5 μM), ATG-F induced strong and stable aggregation of up to 80–100% in a dose-dependent manner (25–100 μg/ml), whereas control IgG did not. The priming effect of ADP was dependent on both P2Y1 and P2Y12 as evidenced by respective ADP receptor antagonists. Preincubation of platelets with inhibitory CD32 mAb completely abolished ATG-F-induced platelet aggregation, suggesting that clustering of, and signalling through, FcγRIIA was crucial for this platelet response. Similarly, ATG-F-derived F(ab′)2 had no effect on ADP-triggered platelet aggregation. Furthermore, preliminary studies indicated that responsiveness of platelet donors to ATG-F was associated with the FcγRIIA-R/R131 polymorphism, which has previously been shown to confer increased in vitro platelet responsiveness to heparin/PF4 antibodies from patients with HIT. In a 14C-serotonin release assay, in which prolonged incubation at 370C under shaking conditions increases baseline platelet activation and ensures constant platelet-platelet contacts, ATG-F (100 μg/ml), but not control IgG, induced strong FcγRIIA-dependent dense-granule release of up to 80%. In contrast, using diluted platelet-rich plasma under static conditions, ATG-F had only minor effects on platelet activation, increasing CD62P+ platelets from 2±1 to 15±5% (n=5). In a cohort of 16 consecutive HSCT patients receiving ATG-F, two patients were identified with normal pretreatment platelet counts and soluble CD62P (sCD62P) plasma levels of >51 ng/ml (mean+2SD of 23 controls). In these patients, an ATG-F-induced drop in platelet count to 38 and 19% of baseline was associated with a 1.7 and 2.2fold increase in sCD62P, respectively, indicating further platelet activation. In summary, strong binding of ATG-F to resting platelets may accelerate their clearance by the reticulo-endothelial system, thus contributing to the pathophysiology of thrombocytopenia. While ATG-F alone had negligible effects on resting platelets, it significantly enhanced activation of prestimulated platelets. Patients with normal-to-high platelet counts and evidence of in vivo platelet activation may especially be prone to this potentially hazardous side effect.

2018 ◽  
Vol 115 (11) ◽  
pp. 1672-1679 ◽  
Author(s):  
Qi Ma ◽  
Weilin Zhang ◽  
Chongzhuo Zhu ◽  
Junling Liu ◽  
Quan Chen

Abstract Aims AKT kinase is vital for regulating signal transduction in platelet aggregation. We previously found that mitochondrial protein FUNDC2 mediates phosphoinositide 3-kinase (PI3K)/phosphatidylinositol-3,4,5-trisphosphate (PIP3)-dependent AKT phosphorylation and regulates platelet apoptosis. The aim of this study was to evaluate the role of FUNDC2 in platelet activation and aggregation. Methods and results We demonstrated that FUNDC2 deficiency diminished platelet aggregation in response to a variety of agonists, including adenosine 5′-diphosphate (ADP), collagen, ristocetin/VWF, and thrombin. Consistently, in vivo assays of tail bleeding and thrombus formation showed that FUNDC2-knockout mice displayed deficiency in haemostasis and thrombosis. Mechanistically, FUNDC2 deficiency impairs the phosphorylation of AKT and downstream GSK-3β in a PI3K-dependent manner. Moreover, cGMP also plays an important role in FUNDC2/AKT-mediated platelet activation. This FUNDC2/AKT/GSK-3β/cGMP axis also regulates clot retraction of platelet-rich plasma. Conclusion FUNDC2 positively regulates platelet functions via AKT/GSK-3β/cGMP signalling pathways, which provides new insight for platelet-related diseases.


2010 ◽  
Vol 78 (10) ◽  
pp. 4268-4275 ◽  
Author(s):  
Oonagh Shannon ◽  
Matthias Mörgelin ◽  
Magnus Rasmussen

ABSTRACT The Gram-positive bacterium Aerococcus urinae can cause infectious endocarditis (IE) in older persons. Biofilm formation and platelet aggregation are believed to contribute to bacterial virulence in IE. Five A. urinae isolates from human blood were shown to form biofilms in vitro, and biofilm formation was enhanced by the presence of human plasma. Four of the A. urinae isolates caused platelet aggregation in platelet-rich plasma from healthy donors. The Au3 isolate, which induced platelet aggregation in all donors, also activated platelets, as determined by flow cytometry. Platelet aggregation was dependent on bacterial protein structures and on platelet activation since it was sensitive to both trypsin and prostaglandin E1. Plasma proteins at the bacterial surface were needed for platelet aggregation; and roles of the complement system, fibrinogen, and immunoglobulin G were demonstrated. Complement-depleted serum was unable to support platelet aggregation by Au3 and complement blockade using compstatin-inhibited platelet activation. Platelet activation by Au3 was inhibited by blocking of the platelet fibrinogen receptor, and this isolate was also shown to bind to radiolabeled fibrinogen. Removal of IgG from platelet-rich plasma by a specific protease inhibited the platelet aggregation induced by A. urinae, and blockade of the platelet FcRγIIa hindered platelet activation induced by Au3. Convalescent-phase serum from a patient with A. urinae IE transferred the ability of the bacterium to aggregate platelets in an otherwise nonresponsive donor. Our results show that A. urinae exhibits virulence strategies of importance for IE.


Toxins ◽  
2020 ◽  
Vol 12 (7) ◽  
pp. 426
Author(s):  
Yu-Ju Kuo ◽  
Yao Tsung Chang ◽  
Ching-Hu Chung ◽  
Woei-Jer Chuang ◽  
Tur-Fu Huang

Polymer polyethylene glycol (PEG), or PEGylation of polypeptides improves protein drug stability by decreasing degradation and reducing renal clearance. To produce a pharmaceutical disintegrin derivative, the N-terminal PEGylation technique was used to modify the disintegrin derivative [KGDRR]trimucrin for favorable safety, pharmacokinetic profiles, and antithrombotic efficacy. We compared intact [KGDRR]trimucrin (RR) and PEGylated KGDRR (PEG-RR) by in vitro and in vivo systems for their antithrombotic activities. The activity of platelet aggregation inhibition and the bleeding tendency side effect were also investigated. PEG-RR exhibited optimal potency in inhibiting platelet aggregation of human/mouse platelet-rich plasma activated by collagen or ADP with a lower IC50 than the intact derivative RR. In the illumination-induced mesenteric venous thrombosis model, RR and PEG-RR efficaciously prevented occlusive thrombosis in a dose-dependent manner. In rotational thromboelastometry assay, PEG-RR did not induce hypocoagulation in human whole blood even given at a higher concentration (30 μg/mL), while RR slightly prolonged clotting time. However, RR and PEG-RR were not associated with severe thrombocytopenia or bleeding in FcγRIIa-transgenic mice at equally efficacious antithrombotic dosages. We also found the in vivo half-life of PEGylation was longer than RR (RR: 15.65 h vs. PEG-RR: 20.45 h). In conclusion, injectable PEG-RR with prolonged half-life and decreased bleeding risk is a safer anti-thrombotic agent for long-acting treatment of thrombus diseases.


1967 ◽  
Vol 17 (01/02) ◽  
pp. 078-098 ◽  
Author(s):  
M. I Barnhart ◽  
D. C Cress ◽  
R. L Henry ◽  
J. M Riddle

SummaryBreakdown products of fibrinogen and fibrin can play a role in hemostasis and also may be of consequence in thrombosis. β2 fibrinogen derivative D is an electropositive terminal proteolysis product of fibrinolysis which has the ability to aggregate platelets. The normal plasma concentration of such nonclottable fibrinogen relatives is 0.2 mg/ml. During fibrinolysis this concentration may reach 5 mg/ml plasma. Addition of β 2 fibrinogen D (raising the plasma concentration 0.1 to 5 mg/ml) either in vivo or in vitro induced platelet aggregations. Moreover, alterations in platelet morphology occurred which were obvious by electron microscopy.Platelet depletion was a consistent response to the infusion of purified β2 fibrinogen D (8 to 55 mg/kg body weight) into dogs. Circulating platelets decreased as much as 85% but were only temporarily aggregated and reappeared in the circulation within 1 to 5 hrs. Small platelet aggregates circulated while large aggregates were trapped in the microcirculation. Thrombin was not responsible for the platelet aggregations as neither prothrombin nor clottable fibrinogen were changed significantly. The transient nature and morphological features of the platelet response according to microscopic criteria were prominent during and after infusion of β2 fibrinogen D.In vitro studies included 3 systems; washed platelets, platelet rich plasma and whole blood. Positive results were obtained with all, but platelets in whole blood were most responsive. The magnitude of platelet aggregation and morphology correlated with the concentration of β2 fibrinogen D. Platelet aggregation induced by ADP (10~5 mg/ml) was compared with that induced by β2 fibrinogen D (0.09 to 0.72 mg/ml). With either reagent aggregates were of dendritic forms. Combination of the 2 reagents was additive but did not further change the morphology. Additional factors seem necessary for development of viscous metamorphosis.


1985 ◽  
Vol 54 (02) ◽  
pp. 480-484 ◽  
Author(s):  
I A Greer ◽  
J J Walker ◽  
M McLaren ◽  
A A Calder ◽  
C D Forbes

SummaryPlatelet aggregation and thromboxane A2 have been implicated in the pathogenesis of several forms of vascular disease. The aim of this study was to determine the effect of a wide range of adrenoceptor antagonists on platelet aggregation, and thromboxane A2 production, from normal human platelet rich plasma in vitro. Labetalol, pindolol and propranolol inhibited platelet aggregation to collagen in a dose dependent manner. Increasing the concentration of collagen “shifted” the dose response curve to the right. These 3 drugs also significantly inhibited thromboxane A2 generation in response to collagen but not to arachidonic acid. This effect was independent of any inhibitory effect of these drugs on platelet aggregation, and occurred at a drug concentration close to that obtained in vivo. Atenolol, metoprolol, prazosin and timolol were similarly assessed but had no effect on either platelet aggregation or thromboxane A2 generation. This ability of labetalol, pindolol, and propranolol to inhibit platelet aggregation and thromboxane generation, may be of clinical benefit in view of the increasing evidence implicating thromboxane A2 in the pathogenesis of vascular disease.


1985 ◽  
Vol 53 (03) ◽  
pp. 337-342 ◽  
Author(s):  
S Krishnamurthi ◽  
V V Kakkar

SummaryThe effect of pyridoxal 5’-phosphate (PALP) and trifluoperazine (TFPZ), the calmodulin antagonist, on in vitro platelet adhesion to collagen and collagen-induced platelet activation was studied using platelet-rich-plasma (PRP) or washed platelets (WPL). Platelet aggregation and [14C]-5HT release induced by “threshold” or low concentrations of collagen (0.6 μg/ ml) in PRP were completely abolished by PALP (24 mM), TFPZ (250 μM) as well as indomethacin (10 μM). At higher concentrations of collagen (10–15 μg/ml) in PRP and WPL, the use of stirred and unstirred platelets treated with collagen enabled a distinction to be made between aggregation and adhesion- mediated release reaction. Platelet aggregation and the aggregation-mediated release reaction induced by these concentrations of collagen in stirred platelets were completely abolished by PALP, TFPZ and indomethacin although neither adhesion to collagen nor the adhesion-mediated release reaction of unstirred platelets was significantly affected by these inhibitors. Interestingly, both adhesion and the adhesion-mediated release reaction were abolished by concentrations of PALP 10–40 fold higher than those required to abolish aggregation. Collagen-induced platelet aggregation, but not platelet adhesion, was inhibited in resuspended platelets pretreated with PALP and NaBH4 indicating a separation in the membrane sites involved in aggregation and adhesion. The results further emphasize the distinction between adhesion and aggregation-mediated events with regards to collagen with the latter being more susceptible to inhibition by antiplatelet agents such as PALP and TFPZ.


1981 ◽  
Author(s):  
M M Frojmovie ◽  
J G Milton ◽  
S S Tang

MPS is characterized by autosomal dominant inheritance thrombocytopenia (5,000-120,000 μl-1), abnormally large platelets on peripheral blood smear, spontaneous aggregation (SA), normal clot retraction and thromboplastin formation, nypervolumetric shape change and normal amounts of plasma membrane. Here we describe further characteristics of MPS for 3 siblings /5 affected family members. The ultrastructural features of MPS platelets were normal except for the high frequency of giant granules. Platelet aggregation (PA) was studied in citrated platelet-rich plasma at platelet counts of 5,000-40,000 μl-1 and was quantitated microscopically from the decrease in single platelet count. PA could be increased over that due to SA alone by the addition of ADP, adrenalin, collagen, ionophore A23187, arachidonic acid and ristocetin, suggesting that the response to these agents may be normal. The ristocetin-induced increase in PA was completely blocked by an IgG specific for BSS. In contrast, over a 4 year period, thrombin (0.2 units/ml) either did not or only slightly increased PA over SA for 3/3 donors. However, MPS platelets pelleted from ACD-PRP and resuspended in Tyrodes’, pH 7.4 at a platelet count of ~ 200,000 μ1-1 showed a normal response to thrombin by aggregometry. Only one donor’s platelets had both reduced sialic acid and glycoprotein (G.P.) 1 reduction/abnormality with otherwise normal G.P. and protein profile, while the other 2 siblings’ platelets showed no such abnormalities. The above results indicate that biochemical variability exists for MPS platelets from different affected siblings. Moreover, our observations raise the possibility that platelet aggregation abnormalities in thrombobytopenic disorders (may be obscured by (1) preparation artefacts and/or (2) artificially increasing platelet counts for in vitro studies.


1988 ◽  
Vol 74 (5) ◽  
pp. 491-497 ◽  
Author(s):  
D. Sils ◽  
S. E. Rodgers ◽  
J. V. Lloyd ◽  
K. M. Wilson ◽  
D. M. Siebert ◽  
...  

1. The aspirin concentrations previously reported to inhibit platelet aggregation in vitro (40–500 μmol/l) are much greater than those required in vivo in man (5 μmol/l). 2. Human platelet-rich plasma was incubated with buffer or various aspirin concentrations at 37°C for up to 4.5 h. Platelet aggregation and thromboxane generation were measured in response to collagen (0.4–6.3 μg/ml) and adenosine 5′-pyrophosphate (0.5–4 μmol/l). 3. The concentration of aspirin needed to inhibit platelet aggregation in response to a critical concentration of aggregating agent (lowest concentration to cause greater than 50% aggregation) was lower than that required for higher concentrations of aggregating agent. 4. With more prolonged incubation times with aspirin, lower concentrations of aspirin inhibited platelet aggregation. 5. Inhibition of platelet aggregation and thromboxane formation by 10 μmol/l aspirin was maximal by 90 min. There was progressive inhibition by 3 μmol/l aspirin during incubation for 270 min. By the end of this time there was also significant inhibition by 1 μmol/l aspirin. 6. The apparent discrepancy between inhibitory aspirin concentrations in vivo and those observed in vitro in previous studies appears to have been resolved by extending the incubation time of platelets with low aspirin concentrations, thus mimicking the conditions in vivo.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 415-415
Author(s):  
Peter J. Lenting ◽  
Janine J. Hulstein ◽  
Bas de Laat ◽  
Ronald H. Derksen ◽  
Rob Fijnheer ◽  
...  

Abstract Patients with the antiphospholipid syndrome are characterized by the association of thrombosis or pregnancy morbidity with the presence of antibodies against phospholipid-binding proteins, such as beta2-Glycoprotein (beta2-GPI) or prothrombin. In particular, antibodies against beta2-GPI strongly correlate with thrombotic complications. One model that explains this correlation involves an antibody-induced gain-of-function of beta2-GPI, which results in prothrombotic properties of this plasma protein. In an alternative model, beta2-GPI may display anti-thrombotic properties that disappear in an antibody-dependent manner. Of course, both possibilities may occur simultaneously. It should be noted, however, that little is known about the physiological function of beta2-GPI. Several in vitro-based studies have pointed to beta2-GPI as a potential inhibitor of fibrinolysis and/or the intrinsic pathway of coagulation. In the present study, we investigated the hypothesis that beta2-GPI affects platelet function. Addition or immune-depletion of beta2-GPI from platelet-rich plasma had little effect on ADP- or collagen-induced platelet aggregation, whereas it did modulate ristocetin-induced platelet aggregation. In a system employing washed platelets, beta2-GPI completely inhibited platelet aggregation induced by VWF/ristocetin or by a recombinant VWF type 2B mutant (VWF/R1306Q). This suggests that beta2-GPI is directed to the von Willebrand (VWF)-glycoprotein Ib axis. Indeed, beta2-GPI interfered with platelet adhesion to VWF-coated coverslips under conditions of flow as well, resulting in a 2-fold reduction of platelet coverage. In direct binding studies, wt-VWF displayed weak binding to immobilized beta2-GPI. However, conversion of latent VWF into a platelet-binding conformation (either via ristocetin or via a type 2B mutation) induced efficient, dose-dependent and saturable binding of VWF to beta2-GPI. By using a number of recombinant deletion-mutants and individual domains, we found that binding was mediated by the VWF A1 domain. Interestingly, anti-beta2-GPI antibodies isolated from patients with the antiphospholipid syndrome not only interfered with the interaction between beta2-GPI and VWF, but also neutralized the inhibitory effect of beta2-GPI towards VWF-dependent platelet aggregation. Finally, we analysed plasma of antiphospholid-syndrome patients for the presence of VWF that is in its platelet-binding conformation by using a specific nanobody (Hulstein et al (2005) Blood106:3035). Levels of active VWF were increased 1.7-fold in patients compared to healthy individuals or patients that lack beta2-GPI-directed antibodies (p&lt;0.0001). This increase in active VWF is similar to that observed in patients with acquired TTP. In conclusion, we demonstrate that beta2-GPI interacts preferentially with the platelet-binding conformation of VWF, thereby inhibiting VWF-dependent platelet adhesion and aggregation. Anti-beta2-GPI antibodies obtained from antiphospholipid-syndrome patients reverse this anti-thrombotic effect of beta2-GPI. We propose that beta2-GPI functions as a natural first-line barrier that prevents small amounts of active VWF to interact with platelets.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3200-3200
Author(s):  
Todd Meyer ◽  
Liza Robles-Carrillo ◽  
Hina Desai ◽  
Meghan Hatfield ◽  
Mildred Amaya ◽  
...  

Abstract Abstract 3200 Anti-angiogenic therapy with the monoclonal antibody (mAb) bevacizumab (bev; Genentech) is associated with venous and arterial thrombosis in cancer patients. We recently showed that bev immune complexes (ICs) activate platelets in vitro and cause thrombotic thrombocytopenia in mice transgenic for the human platelet IgG receptor, FcγRIIa (FCGR2A mice; Jax Labs). Previous studies conducted in other labs using wild type mice injected with bevacizumab failed to identify thrombotic side effects; however, wild type mice do not have platelet FcγRIIa IgG receptors. Platelets from human donors or from FCGR2A mice are directly activated when FcγRIIa is clustered by the Fc domains of mAbs that have been assembled into higher order ICs by multivalent antigens, such as was shown for bev (Meyer, et al, J Thromb Haemost, 2009;7:171) and for anti-CD154 ICs (Robles-Carrillo, et al, J Immunol, 2010;185:1577). Aflibercept (aflib, Regeneron/sanofi-aventis, also known as VEGF Trap) is a soluble decoy receptor protein that binds all isoforms of VEGF-A including VEGF165 and inhibits VEGF165 activity. Aflib also binds the related angiogenic molecule PlGF, and like bev has anti-angiogenic activity. The aflib molecule contains the Fc domain of the IgG isotype antibody; however, although VEGF165 efficiently clusters bev into higher order ICs, VEGF165 does not cluster aflib into higher order complexes (Rudge JS, et al, PNAS, 2007;104:18363). This suggests that aflib+VEGF165 should not be able to trigger platelet FcγRIIa activation (because FcγRIIa requires clustering for activation). We therefore hypothesized that aflib+VEGF165 complexes would fail to activate platelet FcγRIIa in vitro or in FCGR2A transgenic mice. For in vitro studies, we chose the washed platelet aggregation method as a measure of IC-induced platelet activation. To assess platelet activation in vivo, we measured IC-induced reductions in the number of circulating platelets (thrombocytopenia) following intravenous injection of ICs into FCGR2A mice. Aflib+VEGF165 or bev+VEGF165 complexes were pre-formed at room temperature in the presence or absence of heparin. (Heparin can enhance bev IC-induced platelet activation. Many cancer patients are routinely exposed to heparin to maintain patency of chemotherapy ports.) In 5 donors, bev+VEGF165 complexes (500nM) rapidly induced full aggregation of human platelets in an FcγRIIa-dependent manner, whereas 500nM aflib+VEGF165 failed to induce aggregation under all conditions tested. Within 5 minutes following intravenous injection of bev+VEGF165, FCGR2A mice (n=10) experienced signs of distress consistent with thrombotic shock, including rapid shallow breathing, hunched posture, and decreased or dysfunctional locomotor activity. Animals receiving aflib complexes (n=10) did not exhibit these symptoms. Platelet counts 10 minutes following IC injection revealed thrombocytopenia in animals receiving bev+VEGF165 but not aflib+VEGF165 complexes. Specifically, animals injected with PBS (vehicle; n=5) had baseline platelet counts of (1173±179;mean±SD), whereas animals receiving bev+VEGF165 had mean platelet counts of 331±217 (with heparin; P < 0.001) and 725±303 (without heparin; P < 0.003). Animals receiving aflib+VEGF165 had mean platelet counts of 966±168 (with heparin) and 878±250 (without heparin), which were not statistically different from baseline animal counts. These results indicate that aflibercept lacks that platelet activating potential observed with bevacizumab, and also suggest that anti-angiogenic therapy with aflibercept may lack the capacity to induce the platelet associated thrombotic side effects observed in patients receiving bevacizumab. Disclosures: Meyer: Regeneron Pharmaceuticals, Inc.: Florida Hospital received research funding from Regeneron Pharmaceuticals in support of some of this work.


Sign in / Sign up

Export Citation Format

Share Document