Extracellular Hemoglobin Increases the Binding of the Active Form of Von Willebrand Factor to Glycoprotein Ibα and Platelet Activation Under Shear Stress: Implications for Sickle Cell Disease and Other Hemolytic Disorders

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2095-2095
Author(s):  
Miguel A. Cruz ◽  
Prasenjit Guchhait ◽  
Ryanne Ashley Brown

Abstract Abstract 2095 Sickle cell disease (SCD) is characterized by a hypercoagulable state that accelerates vaso-occlusive events in microcirculation, leading to acute and chronic organ damage. Studies have indicated that excessive release of hemoglobin from erythrocytes into plasma may contribute to platelet activation and thrombosis. This finding is likely due to the infusion of extracellular hemoglobin (ECHb), which causes platelet aggregation and adhesion on prothrombotic surfaces, and inhibits the metalloprotease ADAMTS13, the enzyme that reduces the size of the constitutively active ultra large (UL) von Willebrand factor (VWF) multimers into a smaller non-active form of VWF. In fact, we have described that the binding of ECHb to the A2 domain of VWF prevents the cleavage of VWF by ADAMTS13. This inhibition from ECHb may raise the levels of hyperreactive VWF multimers in blood, contributing to thrombosis. Interestingly, a recent study from another research group described a direct correlation between the rate of hemolysis and the levels of circulating hyperreactive VWF multimers in plasma from SCD patients. We further analyzed the functional consequences of ECHb binding to active VWF multimers and are now describing a novel role for ECHb in VWF-mediated thrombosis. With increasing concentrations of ECHb (0 – 600 μg/ml) mixed with a constant concentration of endothelial-derived ULVWF, we observed incremental increases in the binding capacity of ULVWF for platelet GPIbα. Maximal binding capacity was obtained at 200 μg/ml of ECHb. Identical results were obtained with plasma-derived VWF using a low dose of ristocetin to activate VWF. This finding is due to the preferential binding of ECHb to the active form of VWF. The anti-A2 domain monoclonal antibody VP-1 effectively inhibited the upregulated effect of ECHb on VWF-GPIbα binding by blocking the interaction between ECHb and the A2 domain in both ULVWF and plasma VWF. We then tested the effect of ECHb on VWF-mediated platelet activation/adhesion to a surface coated with fibrin(ogen) under high shear rates. This assay required the addition of a low dose of ristocetin (0.15 mg/ml) to whole blood prior to perfusion. The effect of ristocetin on platelet activation/adhesion was effectively blocked with either EDTA or antibodies against GPIbα and αIIbβ3. ECHb (200 μg/ml) or buffer was added to whole blood prior to perfusion over the fibrin(ogen)-coated surface. At a shear rate of 1500s−1, the amount of activated/adhered platelets observed with blood containing ECHb was significantly higher (300%) than that of blood containing buffer. Comparable results were obtained when ristocetin was substituted with a gain-of-function recombinant A1A2A3 mutant (R1450E, 100nM), which exposes the A2 domain and exhibits increased GPIbα-binding activity. The number of adherent platelets from blood containing only ECHb without ristocetin was <5% of that from blood containing only ristocetin. Previously, we have demonstrated that the dissociation of A1 and A2 domains in VWF increases the binding of A1 to GPIbα. Therefore, we speculate that the interaction of ECHb with the exposed A2 domain in active VWF influences the structure of the neighboring A1 domain, provoking A1 to adopt a conformation with a higher binding affinity for GPIbα. Furthermore, these results suggest an important role for the interaction between ECHb and hyperreactive VWF in SCD pathology, and most likely other conditions presenting with microangiopathic hemolytic anemia. Disclosures: No relevant conflicts of interest to declare.

2011 ◽  
Vol 07 (02) ◽  
pp. 150
Author(s):  
Zhou Zhou ◽  
Prasenjit Guchhait ◽  
◽  

Elevated levels of ultralarge (UL) von Willebrand factor (VWF) multimers in plasma play an important role in cell adhesion and vascular occlusion in sickle cell disease (SCD). Recently, we have shown that the binding of extracellular hemoglobin (ECHb) to the A2 domain of VWF significantly blocks VWF cleavage by the metalloprotease ADAMTS13in vitro. We speculated that on release from the inflamed endothelium, the VWF multimers maintain the UL structure in plasma if ECHb prevents their cleavage. We observed that a subpopulation of VWF multimers that are bound to ECHb (HbVWF), which accounted for about 14 % of the total VWF in the plasma of SCD patients. The plasma HbVWF level is parallely correlated with the ECHb and VWF-antigen levels. The HbVWF multimers are resistant to the metalloprotease ADAMTS13in vitroand are more adhesive to platelets and collagen compared with their Hb-free counterpart. Therefore, we speculate that the HbVWF, which are probably UL multimers, play an important role in tethering and stably adhering blood cells to the vascular endothelium and culminate in vascular occlusion/thrombosis/strokes in SCD patients. Thus, this article provides a new insight into the molecular pathophysiology of SCD.


Blood ◽  
1998 ◽  
Vol 91 (8) ◽  
pp. 2810-2817 ◽  
Author(s):  
Marc F. Hoylaerts ◽  
Chantal Thys ◽  
Jef Arnout ◽  
Jos Vermylen

A patient with a history of recurrent late fetal loss associated with multiple placental infarcts and cerebrovascular ischemia at the age of 36, followed a year later by a myocardial infarction, was referred for further investigation. Coronary angiography was normal. Antinuclear factor, lupus anticoagulant, anticardiolipin antibodies, and other thrombophilia parameters were negative, but there was moderate hyperthyroidism with positive thyroid peroxidase antibodies. Platelet numbers and von Willebrand factor (vWF) were normal. Her platelets showed spontaneous aggregation that disappeared with aspirin intake. However, aggregation still was induced by low levels of ristocetin (0.3 to 0.5 mg/mL). The low-dose ristocetin aggregation in patient platelet-rich plasma (PRP) was completely blocked by neutralizing antiglycoprotein Ib (GPIb) and anti-vWF antibodies. The monoclonal anti-FcγRII receptor antibody IV.3 inhibited partly, which suggests that PRP aggregation by low-dose ristocetin was elicited by vWF-immunoglobulin (Ig) complexes. Upon addition to washed human platelets, with vWF (10 μg/mL), purified patient Igs dose-dependently enhanced ristocetin (0.15 mg/mL)-induced aggregation between 0 and 500 μg/mL, an effect that disappeared again above 1 mg/mL. Aggregation was dependent on the vWF concentration and was blocked by IV.3 or neutralizing anti-GPIb or anti-vWF antibodies. The spontaneous aggregation of normal platelets resuspended in patient plasma could be inhibited totally by IV.3 and partially by neutralizing anti-GPIb or anti-vWF antibodies. Perfusion with normal anticoagulated blood, enriched with 10% of control or patient plasma, over surfaces coated with vWF showed increased platelet adhesion and activation in the presence of patient antibodies. Treatment of the patient with the antithyroid drug thiamazol and temporary corticosteroids, aspirin, and ticlopidine did not correct the platelet hypersensitivity to ristocetin. These observations suggest that some autoantibodies to vWF may both enhance vWF binding to platelets and cause platelet activation through binding to the FcγRII receptor, and thereby may be responsible for a new form of antibody-mediated thrombosis.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3919-3919
Author(s):  
Zhou Zhou ◽  
Han Hyojeong ◽  
Miguel A. Cruz ◽  
Jose A. Lopez ◽  
Jing-fei Dong ◽  
...  

Abstract One of the hallmark events of sickle cell disease (SCD) is vasoocclusion and episodic pain crisis. Although the mechanism of vascular occlusion is very complicated, processes like thrombosis and thromboembolism have been recognized to play an important role in the development of such clinical manifestation in SCD. Studies have shown that the von Willebrand factor (VWF), especially the ultra-large (UL) multimers play a major role in vasoocclusion, which clearly indicates a possible impairment of the VWF-cleaving metalloproteae ADAMTS-13 in these patients with SCD. In a recent work, indeed we have mentioned that the plasma ADAMTS-13 in patients with SCD having normal antigen level showed 35% less protease activity than the normal. There may be several plasma factors responsible for the acquired deficiency of ADAMTS-13 in SCD. Since, the increasing evidences suggest that the elevated level of extracellular hemoglobin (Hb) in plasma parallely associated with the pathogenesis of SCD, we investigated the effects of extracellular Hb on VWF proteolysis by ADAMTS-13. We observed that purified Hb dose-dependently inhibited the ADAMTS-13 cleavage of recombinant(r) VWF and endothelial ULVWF multimers under static and flow conditions. Hb bound to VWF multimers in a saturation-dependent manner and more potently to the rVWFA2 domain (affinity Kd~24nM), which contains the cleavage site for ADAMTS-13. Hb bound also to the ADAMTS-13 (Kd~65nM), with 2.7 times less affinity than to VWFA2. The bindings were neither calcium-dependent nor affected by haptoglobin. However, it is the Hb-binding to VWF that prevented the substrate from being cleaved by ADAMTS-13. These in vitro findings are consistent with the in vivo observations in patients with SCD. An elevated level of extracellular Hb in plasma was inversely correlated (linear regression, r2 =0.6354) with the low activity of ADAMTS-13 in a cohort of ten adult patients with SCD (mean±SE, Hb 346±138 mg/l; activity 33.3±30%) compared to age and gender-matched normal individuals (n=10; Hb 24±8 mg/l; activity 76.2±16%). The data together suggest that patients with SCD suffer from acquired ADAMTS-13 deficiency, primarily because Hb competitively binds and inhibits the proteolysis of VWF multimers, leading to ULVWF accumulation on vascular endothelium and in circulation. The Hb-VWF interaction may therefore be considered as a therapeutic target for reducing thrombotic and vasoocclusive complications in patients with severe hemolysis such as those with SCD.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3058-3058
Author(s):  
Zhou Zhou ◽  
Hyojeong Han ◽  
Mark M. Udden ◽  
Miguel A. Cruz ◽  
Jing-fei Dong ◽  
...  

Abstract Abstract 3058 Poster Board II-1034 Elevated levels of von Willebrand factor (VWF), especially the ultra-large multimers, play a significant role in the pathogenesis of vascular occlusion in sickle cell disease (SCD) by promoting cell adhesion to the endothelium. Investigating the pathophysiology of vaso-occlusion and thrombosis in SCD, we have recently observed that excessive extracellular-hemoglobin (Hb) in plasma significantly inhibited ADAMTS-13 proteolysis of VWF by binding directly to the enzyme cleavage-site on VWF. Here, we further show that subpopulations of VWF multimers, which are bound to extracellular-Hb, exist in plasma. We have successfully isolated the Hb-bound VWF (HbVWF) multimers from SCD patients' plasma using the Ni-NTA column and quantified by commercial kit. The HbVWF multimers exist in 5 to 6-times less quantity than the Hb-free multimers as measured in SCD patients. Purified HbVWF multimers are mostly uncleavable by recombinant ADAMTS-13 in vitro. These HbVWF multimers are hyper active in agglutinating platelets as detected by ristocetin cofactor (RCof) activity assay, and also hyper adhesive to collagen type-III compared to the Hb-free multimers. The HbVWF multimers exists in about 2-fold more quantity in SCD patients than normal individuals [mean percent level ± SE, 8.1±1.8 (individual mean 6 – 11) vs. 16.6±3 (12 – 21), P <0.001; n=10]. Using another sandwich-ELISA assay we have reexamined the HbVWF levels, which showed a similar pattern as above. Further, the increased level of HbVWF multimers exists parallely with an elevated RCof activity of plasma VWF [mean percent activity ± SE, 100.4±15.1 (78 – 124) vs. 132.9±11.4, (109 – 149), P <0.001] and high extracellular-Hb levels [mean mg/L ± SE, 59±6.5 (42 – 96) vs. 281.5±71.7 (184 – 410), P <0.001] in plasma of SCD patients compared to normal individuals. Therefore, we believe that these hyperactive HbVWF multimers play a crucial role in cell adhesion, vascular occlusion and thrombosis in SCD. Also, we speculate that this mechanism is not only limited in SCD, but also occurred in other pathophysiological conditions associated with severe intravascular hemolysis. Disclosures No relevant conflicts of interest to declare.


2009 ◽  
Vol 101 (06) ◽  
pp. 1070-1077 ◽  
Author(s):  
Zhou Zhou ◽  
Hyojeong Han ◽  
Miguel Cruz ◽  
José López ◽  
Jing-Fei Dong ◽  
...  

SummaryVascular occlusion, thromboembolism and strokes are hallmark events in sickle cell disease (SCD). The von Willebrand factor (VWF), largest adhesive protein in circulation, has been implicated as major component in these processes. In SCD, a high level of extracellular haemoglobin (Hb) in plasma has been shown parallely associated with the disease pathogenesis. Investigating the effect of Hb we observed that purified Hb significantly inhibited the ADAMTS-13 cleavage of VWF under static and flow conditions. Hb bound potently to VWF specifically VWFA2 in a saturation-dependent manner with half-maximal binding 24 nM. Inversely, VWFA2 also bound potently to Hb and binding was inhibited by VP1 antibody, which binds to ADAMTS-13 cleavage site on VWF. Microscopic observation also shows that Hb bound specifically to endothelial VWF under flow. Furthermore, the Hb-bound VWF multimers were isolated from plasma. Though, Hb bound also to ADAMTS-13, it is the Hb binding to VWFA2 that prevented the substrate being cleaved by ADAMTS-13. In an observation in a small pool of patients with SCD, high Hb in plasma was inversely correlated with low proteolytic activity of ADAMTS-13. Thus, the observations suggest that the patients with SCD suffer from an acquired ADAMTS-13 deficiency primarily because Hb competitively bound and blocked the proteolysis of VWF, leading to the accumulation of ultra-large VWF multimers in circulation and on endothelium. Therefore, the Hb-VWF interaction may be considered as a therapeutic target for treating thrombotic and vaso-occlusive complications in patients with severe intravascular haemolysis such as those with SCD.


Sign in / Sign up

Export Citation Format

Share Document