scholarly journals Analysis of the role of von Willebrand factor, platelet glycoprotein VI-, and α2β1-mediated collagen binding in thrombus formation

Blood ◽  
2014 ◽  
Vol 124 (11) ◽  
pp. 1799-1807 ◽  
Author(s):  
Yasuaki Shida ◽  
Natalia Rydz ◽  
David Stegner ◽  
Christine Brown ◽  
Jeffrey Mewburn ◽  
...  

Key PointsThe robustness of the VWF:collagen-binding assay is confirmed in a comprehensive evaluation of VWD collagen-binding defects. Collagen binding by VWF, GPVI, and α2β1 have major albeit overlapping functions in primary hemostasis.

Blood ◽  
2010 ◽  
Vol 115 (24) ◽  
pp. 5069-5079 ◽  
Author(s):  
Nicholas Pugh ◽  
Anna M. C. Simpson ◽  
Peter A. Smethurst ◽  
Philip G. de Groot ◽  
Nicolas Raynal ◽  
...  

AbstractExposed subendothelial collagen acts as a substrate for platelet adhesion and thrombus formation after vascular injury. Synthetic collagen-derived triple-helical peptides, designated collagen-related peptide (CRP), GFOGER, and VWF-III, can specifically engage the platelet collagen receptors, glycoprotein VI and integrin α2β1, and plasma von Willebrand factor (VWF), respectively. Hitherto, the role of these 3 collagen-binding axes has been studied indirectly. Use of these uniform peptide substrates, rather than collagen fibers, provides independent control of each axis. Here, we use confocal imaging and novel image analysis techniques to investigate the effects of receptor-ligand engagement on platelet binding and activation during thrombus formation under flow conditions. At low shear (100s−1 and 300s−1), both GFOGER and CRP are required for thrombus formation. At 1000s−1, a combination of either CRP or GFOGER with VWF-III induces comparable thrombus formation, and VWF-III increases thrombus deposition at all shear rates, being indispensable at 3000s−1. A combination of CRP and VWF-III is sufficient to support extensive platelet deposition at 3000s−1, with slight additional effect of GFOGER. Measurement of thrombus height after specific receptor blockade or use of altered proportions of peptides indicates a signaling rather than adhesive role for glycoprotein VI, and primarily adhesive roles for both α2β1 and the VWF axis.


Author(s):  
Gina Perrella ◽  
Jingnan Huang ◽  
Isabella Provenzale ◽  
Frauke Swieringa ◽  
Floor C.J.I. Heubel-Moenen ◽  
...  

Objective: Fibrin is considered to strengthen thrombus formation via integrin αIIbβ3, but recent findings indicate that fibrin can also act as ligand for platelet glycoprotein VI. Approach and Results: To investigate the thrombus-forming potential of fibrin and the roles of platelet receptors herein, we generated a range of immobilized fibrin surfaces, some of which were cross-linked with factor XIIIa and contained VWF-BP (von Willebrand factor-binding peptide). Multicolor microfluidics assays with whole-blood flowed at high shear rate (1000 s −1 ) indicated that the fibrin surfaces, regardless of the presence of factor XIIIa or VWF-BP, supported platelet adhesion and activation (P-selectin expression), but only microthrombi were formed consisting of bilayers of platelets. Fibrinogen surfaces produced similar microthrombi. Markedly, tiggering of coagulation with tissue factor or blocking of thrombin no more than moderately affected the fibrin-induced microthrombus formation. Absence of αIIbβ3 in Glanzmann thrombasthenia annulled platelet adhesion. Blocking of glycoprotein VI with Fab 9O12 substantially, but incompletely reduced platelet secretion, Ca 2+ signaling and aggregation, while inhibition of Syk further reduced these responses. In platelet suspension, glycoprotein VI blockage or Syk inhibition prevented fibrin-induced platelet aggregation. Microthrombi on fibrin surfaces triggered only minimal thrombin generation, in spite of thrombin binding to the fibrin fibers. Conclusions: Together, these results indicate that fibrin fibers, regardless of their way of formation, act as a consolidating surface in microthrombus formation via nonredundant roles of platelet glycoprotein VI and integrin αIIbβ3 through signaling via Syk and low-level Ca 2+ rises.


1987 ◽  
Author(s):  
M Yamamoto ◽  
Y Ando ◽  
K Watanabe ◽  
H Iri ◽  
Y Araki ◽  
...  

Recently it has been reported that, in addition to binding to glycoprotein (GP) lb, vWF also interacts with GPIIb/IIIa, although the physiological relevance of this interaction is not completeley clear. In this paper, we have investigated the role of different size of vWF multimers in vWF-mediated platelet aggregation. Different size of vWF multimers were purified from human plasma through Sephacryl S-1000 column according to the method of Fowler et al. Fractions were analysed by SDS-agarose gel electrophoresis by the method of Ruggeri et al. When each fraction was examined for ristocetin cofactor activity (RCo), only larger multimers exhibited significant RCo. The maximum extent of ristocetin-induced platelet agglutination by larger multimers (10 μg/ml) was 80%, while that of intermediate and lower multimers at the same concentration was 20% and 0%, respectively. Each fraction was then added to washed platelet suspensions in the presence of 10 μM ADP and 0.3 mM CaCl2. Only larger multimers induced platelet aggregation, while intermediate and lower multimers failed to induce platelet aggregation. The maximum extent of aggregation in the presence of larger multimers (10 μg/ml) was 70% of that in the presence of fibrinogen instead. Similar experiments were peformed using platelet-rich plasma from a patient with afibrinogenemia in stead of washed normal platelets. ADP caused significant aggregation only when purified vWF larger multimers or fibrinogen was added. This vWF-mediated aggregation was completely inhibited by monoclonal antibody to GPIIb/IIIa (1 μg/ml) and synthetic peptide, Arg-Gly-Asp-Ser, (1 mM).Our results indicate that larger multimers of vWF play major roles in vWF interaction with GPIIb/IIIa.


2020 ◽  
Vol 4 (13) ◽  
pp. 2953-2961 ◽  
Author(s):  
Magdolna Nagy ◽  
Gina Perrella ◽  
Amanda Dalby ◽  
M. Francisca Becerra ◽  
Lourdes Garcia Quintanilla ◽  
...  

Abstract The role of glycoprotein VI (GPVI) in platelets was investigated in 3 families bearing an insertion within the GP6 gene that introduces a premature stop codon prior to the transmembrane domain, leading to expression of a truncated protein in the cytoplasm devoid of the transmembrane region. Western blotting and flow cytometry of GP6hom (homozygous) platelets confirmed loss of the full protein. The level of the Fc receptor γ-chain, which associates with GPVI in the membrane, was partially reduced, but expression of other receptors and signaling proteins was not altered. Spreading of platelets on collagen and von Willebrand factor (which supports partial spreading) was abolished in GP6hom platelets, and spreading on uncoated glass was reduced. Anticoagulated whole blood flowed over immobilized collagen or a mixture of von Willebrand factor, laminin, and rhodocytin (noncollagen surface) generated stable platelet aggregates that express phosphatidylserine (PS). Both responses were blocked on the 2 surfaces in GP6hom individuals, but adhesion was not altered. Thrombin generation was partially reduced in GP6hom blood. The frequency of the GP6het (heterozygous) variant in a representative sample of the Chilean population (1212 donors) is 2.9%, indicating that there are ∼4000 GP6hom individuals in Chile. These results demonstrate that GPVI supports aggregation and PS exposure under flow on collagen and noncollagen surfaces, but not adhesion. The retention of adhesion may contribute to the mild bleeding diathesis of GP6hom patients and account for why so few of the estimated 4000 GP6hom individuals in Chile have been identified.


1994 ◽  
Vol 86 (2) ◽  
pp. 327-332 ◽  
Author(s):  
Edith Fressinaud ◽  
Augusto B. Federici ◽  
Giancarlo Castaman ◽  
Chantal Rothschild ◽  
Francesco Rodeghiero ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 29-29
Author(s):  
Veronica H. Flood ◽  
Abraham C. Schlauderaff ◽  
Paula M. Jacobi ◽  
Tricia L. Slobodianuk ◽  
Robert R. Montgomery ◽  
...  

Abstract Von Willebrand factor (VWF) plays a key role in coagulation by tethering platelets to injured subendothelium via binding sites for platelet glycoprotein Ib and collagen. The binding sites for types I and III collagen in the VWF A3 domain are well characterized, and defects in this region have been implicated in von Willebrand disease (VWD). Additional collagens present in the vasculature may also be involved in interactions with VWF. A VWF A1 sequence variation, p.R1399H, has been associated with decreased binding to type VI collagen, but the clinical significance of this observation remains unclear. Type IV collagen is a common component of the basement membrane and as such may be an important ligand for VWF. While some VWD testing utilizes types I or III collagen, current clinical testing does not include collagen IV or VI. To characterize the role of the VWF A1 domain in VWF-type IV collagen interactions, we generated several A1 domain variant human and/or murine recombinant VWF (rVWF) constructs including R1399H and several type 2M VWD variants localized to the same region (S1387I, Q1402P, and an 11 amino acid deletion mutant encompassing amino acids 1392-1402). These constructs were then expressed in HEK 293T cells. To further assess the role of the A1 domain, scanning alanine mutagenesis (SAM) of residues 1387 through 1412 was conducted. VWF antigen levels (VWF:Ag), collagen binding with type III (VWF:CB3), IV (VWF:CB4), or VI (VWF:CB6) collagen were determined, and multimer distribution was assessed for all recombinant VWF variants. The role of R1399H in the context of human rVWF was characterized initially. Although VWF:Ag, VWF:CB3, and multimer distribution were normal for R1399H compared to wild-type (WT VWF), VWF:CB4 was undetectable. To examine this effect in a mouse model, the R1399H variant was expressed in the context of murine rVWF and collagen binding was determined. Similar to the human variant, murine R1399H rVWF demonstrated significantly reduced binding to murine type IV collagen, at only 7% of the binding seen with WT murine rVWF. In order to examine the behavior of R1399H under shear conditions, either WT or R1399H murine rVWF DNA was hydrodynamically injected into the tail veins of VWF -/- mice to induce expression of the proteins; blood was drawn from the vena cava 24 hours later and then examined on the VenaFlux flow apparatus. VWF expression levels and multimer distribution were similar for the R1399H- and WT-injected mice. Under static conditions, the murine plasma-derived R1399H demonstrated decreased VWF:CB4, at only 16% of the levels seen with WT VWF. No defect was seen in VWF:CB3. Furthermore, when binding to type IV collagen was assessed under flow conditions by VenaFlux, platelet adhesion was significantly decreased in mice expressing R1399H VWF as compared to mice expressing WT VWF. When examining other A1 domain variants, Q1402P and del1392-1402 demonstrated absent VWF:CB4 while S1387I demonstrated a significant reduction in VWF:CB4 compared to WT VWF. All SAM VWF A1 domain variants demonstrated normal expression, multimerization, and VWF:CB3. However, type IV collagen binding was absent for R1392A, R1395A, R1399A, and K1406A and was reduced to less than 50% of WT VWF for Q1402A, K1405A, and K1407A. These residues map to an outside face of the VWF A1 domain crystal structure, and are likely the critical residues for VWF binding to type IV collagen. Taken together, these data demonstrate that the type IV collagen binding site localizes to a specific region of the VWF A1 domain. Mutations in this region of VWF may be clinically significant due to a defect in the ability of VWF to attract platelets to exposed type IV collagen which may contribute to bleeding symptoms seen in VWD. Disclosures: No relevant conflicts of interest to declare.


2007 ◽  
Vol 98 (12) ◽  
pp. 1285-1290 ◽  
Author(s):  
Lakshmi Wijeyewickrema ◽  
Elizabeth Gardiner ◽  
Masaaki Moroi ◽  
Michael Berndt ◽  
Robert Andrews

SummaryGlycoprotein (GP)VI, that binds collagen, together with GPIb- IX-V which binds von Willebrand factor, forms an adheso-signalling complex on platelets that initiates thrombus formation in haemostasis and thrombosis. In this study, we show that two snake venom metalloproteinases, crotarhagin and alborhagin, induce ectodomain shedding of GPVI by a mechanism that involves activation of endogenous platelet metalloproteinases. Alborhagin is a viper venom metalloproteinase from Trimeresurus albolabris, while crotarhagin is a previously undescribed toxin from the rattlesnake Crotalus horridus horridus (~60-kDa non-reduced and reduced). Like alborhagin, crotarhagin induces aggregation in human platelet-rich plasma (maximal activity, ~0.3 μg/ ml). Aggregation of washed platelets was inhibited by soluble GPVI ectodomain expressed as an Fc-fusion protein, confirming crotarhagin targeted GPVI. Treating washed platelets with crotarhagin or alborhagin resulted in time-dependent loss of surface GPVI and the appearance of an ~55-kDa soluble GPVI fragment in supernatants. Crotarhagin also induced shedding in GPVItransfected RBL-2H3 cells. Crotarhagin-induced shedding was metalloproteinase-dependent (inhibited by EDTA), but also blocked by inhibitors of GPVI signalling (Src kinase inhibitors, PP1 or PP2, or Syk inhibitor, piceatannol), indicating shedding required GPVI-dependent platelet activation. Together, the data suggest that the rattlesnake metalloproteinase, crotarhagin, and the viper toxin alborhagin, induce GPVI shedding by a mechanism involving activation of endogenous platelet metalloproteinases rather than direct cleavage of GPVI.


Blood ◽  
2009 ◽  
Vol 113 (21) ◽  
pp. 5323-5329 ◽  
Author(s):  
Fumiaki Banno ◽  
Anil K. Chauhan ◽  
Koichi Kokame ◽  
Jin Yang ◽  
Shigeki Miyata ◽  
...  

Abstract ADAMTS13 is a multidomain protease that limits platelet thrombogenesis through the cleavage of von Willebrand factor (VWF). We previously identified 2 types of mouse Adamts13 gene: the 129/Sv-strain Adamts13 gene encodes the long-form ADAMTS13 having the same domains as human ADAMTS13, whereas the C57BL/6-strain Adamts13 gene encodes the short-form ADAMTS13 lacking the distal C-terminal domains. To assess the physiologic significance of the distal C-terminal domains of ADAMTS13, we generated and analyzed 129/Sv-genetic background congenic mice (Adamts13S/S) that carry the short-form ADAMTS13. Similar to wild-type 129/Sv mice (Adamts13L/L), Adamts13S/S did not have ultralarge VWF multimers in plasma, in contrast to 129/Sv-genetic background ADAMTS13-deficient mice (Adamts13−/−). However, in vitro thrombogenesis under flow at a shear rate of 5000 s−1 was accelerated in Adamts13S/S compared with Adamts13L/L. Both in vivo thrombus formation in ferric chloride–injured arterioles and thrombocytopenia induced by collagen plus epinephrine challenge were more dramatic in Adamts13S/S than in Adamts13L/L but less than in Adamts13−/−. These results suggested that the C-terminally truncated ADAMTS13 exhibited decreased activity in the cleavage of VWF under high shear rate. Role of the C-terminal domains may become increasingly important under prothrombotic conditions.


Blood ◽  
1989 ◽  
Vol 74 (3) ◽  
pp. 1031-1035 ◽  
Author(s):  
MS Read ◽  
SV Smith ◽  
MA Lamb ◽  
KM Brinkhous

Botrocetin (venom coagglutinin) induces binding of von Willebrand factor (vWF) to platelet glycoprotein Ib (GPIb), resulting in platelet agglutination. A mechanism whereby botrocetin causes vWF to change to an active platelet-agglutinating form is proposed. Incubation of native vWF with botrocetin yielded an increasingly active vWF with slower migration in two-dimensional immunoelectrophoresis but with no apparent change in vWF multimer pattern. The “activated” vWF eluted mainly in the void volume (Vo) (Bio-Gel A-15m column chromatography). Botrocetin eluted in the included volume (Vi). Vo peaks appeared to contain a vWF- botrocetin complex, based on bioassays and immunoassays. 125I- Botrocetin mixed with vWF eluted in two peaks: in the Vo, coincident with active vWF, and in the Vi. With von Willebrand disease (vWD) plasma lacking vWF, 125I-Botrocetin eluted in the Vi only. It did not bind to platelets without vWF. In aggregometric studies, antibodies (Ab) against botrocetin, vWF, and GPIb prevented botrocetin-induced platelet agglutination and caused dissolution of preformed platelet agglutinates. Immunostaining of aggregates with antibotrocetin Ab revealed a positive reaction. Botrocetin appears to act in a two-step manner, first binding with vWF to form a complex, which then binds to GPIb to cause agglutination. All three components, vWF, botrocetin, and GPIb, appear to be required for maintenance of stable platelet agglutinates.


Blood ◽  
1989 ◽  
Vol 74 (3) ◽  
pp. 1031-1035 ◽  
Author(s):  
MS Read ◽  
SV Smith ◽  
MA Lamb ◽  
KM Brinkhous

Abstract Botrocetin (venom coagglutinin) induces binding of von Willebrand factor (vWF) to platelet glycoprotein Ib (GPIb), resulting in platelet agglutination. A mechanism whereby botrocetin causes vWF to change to an active platelet-agglutinating form is proposed. Incubation of native vWF with botrocetin yielded an increasingly active vWF with slower migration in two-dimensional immunoelectrophoresis but with no apparent change in vWF multimer pattern. The “activated” vWF eluted mainly in the void volume (Vo) (Bio-Gel A-15m column chromatography). Botrocetin eluted in the included volume (Vi). Vo peaks appeared to contain a vWF- botrocetin complex, based on bioassays and immunoassays. 125I- Botrocetin mixed with vWF eluted in two peaks: in the Vo, coincident with active vWF, and in the Vi. With von Willebrand disease (vWD) plasma lacking vWF, 125I-Botrocetin eluted in the Vi only. It did not bind to platelets without vWF. In aggregometric studies, antibodies (Ab) against botrocetin, vWF, and GPIb prevented botrocetin-induced platelet agglutination and caused dissolution of preformed platelet agglutinates. Immunostaining of aggregates with antibotrocetin Ab revealed a positive reaction. Botrocetin appears to act in a two-step manner, first binding with vWF to form a complex, which then binds to GPIb to cause agglutination. All three components, vWF, botrocetin, and GPIb, appear to be required for maintenance of stable platelet agglutinates.


Sign in / Sign up

Export Citation Format

Share Document