scholarly journals Autoinhibition of Mouse Von Willebrand Factor: Addressing the Species Compatibility between VWF and Gpibα in Humans and Mice

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1002-1002
Author(s):  
Nicholas A Arce ◽  
Renhao Li

Abstract Introduction: The usage of mouse models to investigate thrombosis and hemostasis has now spanned several decades, with the advent of genetic manipulation elucidating the roles of platelet receptors, secondary messengers, and coagulation factors to name a few. While there are many similarities between humans and mice, the vast differences in physical parameters of blood circulation and coagulation should not be ignored. The incompatibility of mouse platelet receptor glycoprotein (GP)Ibα with human von Willebrand factor (VWF) has been well documented, yet human GPIbα can interact with mouse plasma VWF under shear. In light of the recently discovered autoinhibitory module (AIM) in modulating the activity of human VWF, here we seek to address the role of the AIM in species compatibility between these two interacting proteins. Methods: Recombinant human and mouse VWF fragments containing the A1 domain and either intact or truncated AIM, followed by a poly-histidine tag, were expressed from stably transfected mammalian cells. Recombinant human and mouse GPIbα ligand-binding domain (LBD) (residues 1-290, mature protein numbering) were similarly expressed bearing a biotin acceptor sequence and poly-histidine tag. Stable cell lines co-expressing E. coli biotin ligase were used to express and biotinylate the LBD protein before purification. All proteins were purified by nickel-affinity chromatography followed by size exclusion chromatography. Bio-layer interferometry was performed on an Octet QK e using streptavidin biosensors to capture biotinylated GPIbα LBD fragments. Data was analyzed using a 2:1 heterogeneous ligand binding model for kinetic parameters, and steady state affinity (K D) was calculated either from binding responses at the end of the association phase, or from predicted equilibria from the aforementioned model. Human or mouse washed platelets were isolated by centrifugation, followed by pelleting the PRP in the presence of PGE-1. Platelets were resuspended to 150,000/ μL in modified Tyrode's buffer, and aggregation was monitored in a light transmission aggregometer. Results: Consistent with earlier reports, human AIM-A1 protein (VWF residues 1238-1493), at concentrations below 1 μM, showed no appreciable binding to human GPIbα LBD. Conversely, human truncated AIM-A1 protein (tAIM-A1, residues 1261-1472) exhibited a K D of 348 nM towards the LBD. Mouse tAIM-A1 proteins showed enhanced binding to mouse LBD (K D = 214 nM), but mouse AIM-A1 is able to bind to mouse LBD with a K D of 716 nM, which is considerably higher than the affinity of the human-human pair of tens of μM (Figure 1). Given the increased affinity of mouse AIM-A1 to mouse LBD, we reasoned that the mouse AIM may be less stable than the human counterpart. A chimeric AIM-A1 protein, in which human AIM sequences flank mouse A1 (human residues 1238-1271, mouse 1272-1458, and human 1459-1493), was generated. The chimeric AIM-A1 showed little binding to mouse LBD. However, this protein can still aggregate washed platelets from both species and bind to gain-of-function human GPIbα, indicating it still is a folded domain capable to recognizing its cognate receptor, mouse GPIbα (Figure 2). The isolated mouse AIM-A1 fragments are capable of inducing robust mouse and human platelet aggregation for washed platelets in a dose dependent manner. It had been previously demonstrated that in platelet rich plasma, ristocetin has no effect on mouse VWF. However, washed mouse platelets incubated with 60 nM of mouse AIM-A1 followed by addition of 1.5 mg/mL ristocetin was able to agglutinate the platelets (Figure 2). These results suggest that these proteins may be used to help elucidate the signaling pathways of GPIb-IX-V complex in mice. Conclusion: In agreement with previous reports, we observe that in general mouse AIM-A1 is more active than human AIM-A1 in its propensity to bind to GPIbα. On the aspect that human VWF in incompatible with mouse platelets, we observed that mouse LBD has lower affinity for human tAIM-A1 than human LBD does for mouse tAIM-A1. These findings altogether suggest that there are likely differences in activation forces for each species AIM, and that the GPIbα-A1 interactions, in terms of their mechanical and thermodynamic properties, are somewhat unique, and perhaps optimized, for each species. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 698-698 ◽  
Author(s):  
Thomas A J Mckinnon ◽  
Richard D Starke ◽  
Kushani Ediriwickrema ◽  
Anna Maria Randi ◽  
Michael Laffan

Abstract Abstract 698 Von Willebrand Factor (VWF) is a large multimeric plasma glycoprotein essential for homeostasis, also involved in inflammation and angiogenesis. The majority of VWF is synthesised by endothelial cells (EC) and is either constitutively secreted or stored in Weibel-Palade bodies (WPB), ready to be released in response to endothelial stimulation. Several studies have shown that formation of WPB is dependent on the presence of VWF, and deletion of VWF in human umbilical vein EC (HUVEC) results in loss of WPB. Amongst the other proteins shown to co-localise to WPB is angiopoietin-2 (Ang2), a ligand of the receptor tyrosine kinase Tie-2. Ang2 regulates endothelial cell survival, vascular stability and maturation, by destabilizing quiescent endothelium and facilitating the response to inflammatory and angiogenic stimuli. VWF is required for storage of Ang2, and release of Ang-2 from EC is increased in VWF-deficient HUVEC. Recently, we have shown that VWF itself regulates angiogenesis, raising the hypothesis that some of the angiogenic activity of VWF may be mediated by Ang-2. In the present study we investigated the interaction between Ang2 and VWF. Binding analysis demonstrated that recombinant human Ang2 bound to purified plasma-derived VWF in a pH and calcium dependent manner, with optimal binding occurring at pH 6.5 and 10mM calcium, indicative of binding within the Golgi body. Generation of binding isotherms established that Ang2 bound to VWF with high affinity (KD∼3nM); furthermore binding affinity was not dependent on VWF conformation. Using an array of VWF constructs we determined that Ang2 bound predominantly to the VWF A1 domain, which also contains binding sites to the platelet receptor GPIb and extracellular matrix proteins. Co-immunoprecipitation experiments performed on TNFα- and ionomycin-stimulated HUVECs, to induce WPB exocytosis, confirmed that a portion of Ang2 remained bound to secreted VWF. Moreover, immunofluorescence staining of histamine-stimulated HUVECs to induce VWF release demonstrated the presence of Ang2 on VWF strings secreted from ECs. Finally we demonstrated that Ang2 bound to VWF was still able to interact with Tie-2. These data demonstrate that binding of Ang2 to VWF occurs within the cell; we propose that this is the mechanism mediating storage of Ang2 in WPB. Moreover, the finding that the Ang2-VWF interaction is preserved following secretion raises the intriguing possibility VWF may affect Ang2 function, possibly by localising Ang2 to the Tie 2 receptor under the shear forces experienced in flowing blood. Similarly, Ang-2 binding to VWF may modulate its interaction with receptors and extracellular matrix proteins, and ultimately influence the role of VWF in the angiogenic processes. Disclosures: No relevant conflicts of interest to declare.


1990 ◽  
Vol 64 (04) ◽  
pp. 589-593 ◽  
Author(s):  
Edith Fressinaud ◽  
Jean Pierre Girma ◽  
J Evan Sadler ◽  
Hans R Baumgartner ◽  
Dominique Meyer

SummaryWe compared the effect of a synthetic dodecapeptide of residues 400-411 of the Γ chain of fibrinogen (Γ Fg 400-411) and of three synthetic peptides (15 to 18 aminoacids), of human von Willebrand Factor (vWF), containing the 1744-1747 Arg-Gly-Asp-Ser (RGDS) sequence, upon platelet adhesion to collagen in flowing blood. Both types of peptides are known to inhibit the binding of adhesive proteins to platelet membrane glycoprotein Ilb/IIIa (GPIIb/IIIa). Collagen was coated onto plastic cover slips and exposed in parallel-plate perfusion chambers to reconstituted human blood at various shear rates for 5 min at 37 °C. At a shear rate of 2,600 s−1, RGDS peptides inhibited platelet adhesion to collagen in a dose-dependent manner and appeared to be more potent inhibitors than the Γ Fg 400-411 on a molar basis. No synergetic effect between RGDS and Γ Fg 400-411 peptides was observed. These results suggest that the RGDS peptides affect adhesion by inhibiting the GPIIb/IIIa-vWF interaction and confirm the involvement of this platelet receptor in vWF-mediated platelet adhesion to collagen at high shear rate.


1996 ◽  
Vol 75 (04) ◽  
pp. 655-660 ◽  
Author(s):  
Mario Mazzucato ◽  
Luigi De Marco ◽  
Paola Pradella ◽  
Adriana Masotti ◽  
Francesco I Pareti

SummaryPorcine von Willebrand factor (P-vWF) binds to human platelet glycoprotein (GP) lb and, upon stirring (1500 rpm/min) at 37° C, induces, in a dose-dependent manner, a transmembrane flux of Ca2+ ions and platelet aggregation with an increase in their intracellular concentration. The inhibition of P-vWF binding to GP lb, obtained with anti GP lb monoclonal antibody (LJ-Ib1), inhibits the increase of intracellular Ca2+ concentration ([Ca2+]i) and platelet aggregation. This effect is not observed with LJ-Ib10, an anti GP lb monoclonal antibody which does not inhibit the vWF binding to GP lb. An anti GP Ilb-IIIa monoclonal antibody (LJ-CP8) shown to inhibit the binding of both vWF and fibrinogen to the GP IIb-IIIa complex, had only a slight effect on the [Ca2+]i rise elicited by the addition of P-vWF. No inhibition was also observed with a different anti GP IIb-IIIa monoclonal antibody (LJ-P5), shown to block the binding of vWF and not that of fibrinogen to the GP IIb-IIIa complex. PGE1, apyrase and indomethacin show a minimal effect on [Ca2+]i rise, while EGTA completely blocks it. The GP lb occupancy by recombinant vWF fragment rvWF445-733 completely inhibits the increase of [Ca2+]i and large aggregates formation. Our results suggest that, in analogy to what is seen with human vWF under high shear stress, the binding of P-vWF to platelet GP lb, at low shear stress and through the formation of aggregates of an appropriate size, induces a transmembrane flux of Ca2+, independently from platelet cyclooxy-genase metabolism, perhaps through a receptor dependent calcium channel. The increase in [Ca2+]i may act as an intracellular message and cause the activation of the GP IIb-IIIa complex.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Katarina D. Kovacevic ◽  
Stefan Greisenegger ◽  
Agnes Langer ◽  
Georg Gelbenegger ◽  
Nina Buchtele ◽  
...  

AbstractThe effect of conventional anti-platelet agents is limited in secondary stroke prevention, and their effects are blunted under high shear stress in the presence of increased levels of circulating von Willebrand factor (VWF). VWF is critically involved in thrombus formation at sites of stenotic extracranial/intracranial arteries. A third generation anti-VWF aptamer (BT200) has been generated which could be useful for secondary stroke prevention. To characterize the effects of BT200 in blood of patients with large artery atherosclerosis stroke (LAA). Blood samples were obtained from 33 patients with acute stroke or transient ischemic attack to measure inhibition of VWF activity and VWF-dependent platelet function. Patients who received clopidogrel or dual antiplatelet therapy did not differ in VWF dependent platelet function tests from aspirin treated patients. Of 18 patients receiving clopidogrel with or without aspirin, only 3 had a prolonged collagen adenosine diphosphate closure time, and none of the patients had ristocetin induced aggregation in the target range. BT200 concentration-dependently reduced median VWF activity from 178 to < 3%, ristocetin induced platelet aggregation from 40U to < 10U and prolonged collagen adenosine diphosphate closure times from 93 s to > 300 s. Baseline VWF activity correlated (r = 0.86, p < 0.001) with concentrations needed to reduce VWF activity to < 20% of normal, indicating that BT200 acts in a target concentration-dependent manner. Together with a long half-life supporting once weekly administration, the safety and tolerability observed in an ongoing phase I trial, and the existence of a reversal agent, BT200 is an interesting drug candidate.


Blood ◽  
2019 ◽  
Vol 133 (4) ◽  
pp. 356-365 ◽  
Author(s):  
Reinhard Schneppenheim ◽  
Natalie Hellermann ◽  
Maria A. Brehm ◽  
Ulrike Klemm ◽  
Tobias Obser ◽  
...  

Abstract The frequent von Willebrand factor (VWF) variant p.Phe2561Tyr is located within the C4 domain, which also harbors the platelet GPIIb/IIIa-binding RGD sequence. To investigate its potential effect on hemostasis, we genotyped 865 patients with coronary artery disease (CAD), 915 with myocardial infarction (MI), and 417 control patients (Ludwigshafen Risk and Cardiovascular Health Study) and performed functional studies of this variant. A univariate analysis of male and female carriers of the Tyr2561 allele aged 55 years or younger revealed an elevated risk for repeated MI (odds ratio, 2.53; 95% confidence interval [CI], 1.07-5.98). The odds ratio was even higher in females aged 55 years or younger, at a value of 5.93 (95% CI, 1.12-31.24). Cone and plate aggregometry showed that compared with Phe2561, Tyr2561 was associated with increased platelet aggregate size both in probands’ blood and with the recombinant variants. Microfluidic assays revealed that the critical shear rate for inducing aggregate formation was decreased to 50% by Tyr2561 compared with Phe2561. Differences in C-domain circular dichroism spectra resulting from Tyr2561 suggest an increased shear sensitivity of VWF as a result of altered association of the C domains that disrupts the normal dimer interface. In summary, our data emphasize the functional effect of the VWF C4 domain for VWF-mediated platelet aggregation in a shear-dependent manner and provide the first evidence that a functional variant of VWF plays a role in arterial thromboembolism.


2002 ◽  
Vol 88 (09) ◽  
pp. 421-426 ◽  
Author(s):  
Stefan Lethagen ◽  
Christina Isaksson ◽  
Charlotta Schaedel ◽  
Lars Holmberg

SummaryHereditary defects of the von Willebrand factor (VWF) gene cause von Willebrand’s disease (VWD) which shows great variability dependent on the nature and location of the mutation. We here describe the characteristics of a substitution of methionine for threonine 1156 in the D3 domain of the VWF, i.e. the domain involved in the intracellular multimerization of pro-VWF dimers. A VWD patient with severe symptoms was a compound heterozygote for the T1156M mutation and a null allele (Q2470X) on the other chromosome. This led to marked reduction of plasma VWF concentration to about 0.05 U/ml and an abnormality of VWF multimers as in type 2A VWD. Expression in vitro of the mutation demonstrated that 1156M-VWF is secreted from COS-7 cells in a much reduced amount and lacking large multimers. When coexpressed with normal VWF 1156M-VWF decreased the secretion of normal VWF in a dose-dependent manner, the secreted VWF showing all the multimers. Two relatives of the propositus were single heterozygotes for the T1156M mutation and were either asymptomatic or had the manifestations of mild type 1 VWD. The expression data and studies of platelet VWF indicate that the T1156M mutation results in intracellular retention of VWF rather than impaired synthesis. Three other members of the family were heterozygotes for the Q2470X mutation and demonstrated the variable expressivity of a null allele.


Blood ◽  
1990 ◽  
Vol 75 (3) ◽  
pp. 688-695 ◽  
Author(s):  
EM Paleolog ◽  
DC Crossman ◽  
JH McVey ◽  
JD Pearson

Abstract We examined the effect of cytokines on basal and agonist-stimulated release of von Willebrand factor (vWf) by human endothelial cells. Treatment of endothelial cells for up to 48 hours with human recombinant or purified interleukin 1 (IL-1) or human recombinant tumor necrosis factor-alpha (TNF-alpha) did not significantly affect constitutive secretion of vWf or intracellular levels of vWf, although basal prostacyclin (PGI2) production was markedly enhanced. In contrast, both IL-1 and TNF-alpha modulated vWf release in response to thrombin or phorbol ester. Pretreatment of endothelial cells for 2 hours with either cytokine enhanced by up to threefold the stimulatory effect of a subsequent 60-minute exposure to thrombin. Addition of cycloheximide (5 micrograms/mL) during the preincubation abolished this enhancement. Moreover, if the cytokine pretreatment time was extended to 24 hours, agonist-stimulated vWf release was significantly suppressed. Cytokine treatment for 2 or 24 hours had no detectable effect on levels of vWf messenger RNA. The effects of cytokines were not the result of contamination with bacterial lipopolysaccharide and were not attributable to endothelial cell injury. These results show that cytokines have little or no direct effect on vWf release from endothelial cells but can significantly modulate its acute release in response to other stimuli in a complex time- and dose-dependent manner.


Blood ◽  
1982 ◽  
Vol 59 (3) ◽  
pp. 542-548 ◽  
Author(s):  
HR Gralnick ◽  
MC Cregger ◽  
SB Williams

Abstract The factor VIII/von Willebrand factor (f.VIII/vWf) protein was purified from the plasma of a patient with von Willebrand's disease (vWd). The patient had all of the classic laboratory findings of vWd except for the ristocetin-induced platelet aggregation of his own platelet-rich plasma. The disease has been documented in three generations. Comparison of the purified normal and vWd f.VIIi/vWf protein revealed several abnormalities, including decreased concentration of f.VIII/vWf antigen; decreased specific vWf activity; absence of the larger molecular forms of the f.VIII/vWf protein; carbohydrate deficiencies affecting the sialic acid, penultimate galactose and N- acetylglucosamine moieties; and decreased binding of the f.VIII/vWf protein to its platelet receptor. These studies indicate the multiplicity of biochemical and functional abnormalities associated with the f.VIII/vWf protein in vWd. f.VIII/vWf protein to normal f.VIII/vWf protein that had been treated with 2-mercaptoethanol (2-ME) to reduce the multimer size and then treated with specific exoglycosidases to remove the sialic acid and penultimate galactose residues revealed similar biologic properties.


Blood ◽  
1984 ◽  
Vol 64 (6) ◽  
pp. 1254-1262 ◽  
Author(s):  
H Takahashi ◽  
M Handa ◽  
K Watanabe ◽  
Y Ando ◽  
R Nagayama ◽  
...  

Abstract We studied four patients who showed aggregation of platelets in platelet-rich plasma at lower concentrations of ristocetin than those required for normal platelet-rich plasma and who demonstrated an increased capacity of the platelets to bind normal von Willebrand factor. The four patients were from two Japanese families. Platelets from one family aggregated spontaneously in vitro, and platelets from both families aggregated upon the addition of normal plasma and cryoprecipitate, in the absence of ristocetin or other agonists. Analysis of the multimeric composition of von Willebrand factor by sodium dodecyl sulfate-agarose gel electrophoresis revealed a decrease in large multimers or a decrease in both large and intermediate multimers in plasma, but normal multimers in platelets. 1-Deamino-[8-D- arginine]-vasopressin caused by an immediate appearance of larger multimers in plasma, followed by the rapid disappearance of these multimers from circulating plasma. Analysis of platelet membrane glycoproteins from the patients showed that there were two distinct bands in the glycoprotein I region; one migrated in a slower region and the other in a faster region than normal glycoprotein Ib. We suggest that the platelet receptor abnormality in these patients is related to this abnormality of glycoprotein Ib.


Sign in / Sign up

Export Citation Format

Share Document