scholarly journals Shear-stress-induced von Willebrand factor binding to platelets causes the activation of tyrosine kinase(s)

1994 ◽  
Vol 302 (3) ◽  
pp. 681-686 ◽  
Author(s):  
K Razdan ◽  
J D Hellums ◽  
M H Kroll

Pathological arterial blood flow generates fluid shear stresses that directly cause platelet aggregation. The mechanism of shear-induced platelet aggregation is incompletely understood, but involves von Willebrand factor (vWF) binding to platelet glycoprotein (GP) Ib and GP IIb-IIIa, leading to the transmembrane influx of Ca2+ and the activation of protein kinase C. To investigate this further, shear-stress-induced protein tyrosine phosphorylation (PTP) of washed platelets was studied in a cone-plate viscometer. A time- and shear-stress-dependent tyrosine phosphorylation of substrates with approx. M(r) 29,000-31,000, 36,000, 50,000, 58,000, 64,000, 76,000, 85,000 and 105,000 was observed. PTP in response to a threshold shear stress of 0.3 mN/cm2 (30 dyn/cm2) was enhanced in most cases by exogenous purified human vWF, and PTP in response to a pathological shear stress of 0.9 mN/cm2 (90 dyn/cm2) was inhibited in some cases by inhibiting vWF binding to GP Ib or GP IIb-IIIa, or by inhibiting Ca2+ responses with extracellular EGTA. Shear-induced PTP of a substrate of M(r) approximately 31,000 appeared to be independent of GP Ib, and PTP of a substrate(s) of M(r) approximately 29,000 was shear-stress-dependent but independent of extracellular Ca2+. Cytochalasin D, which inhibits GP Ib-cytoskeleton interactions, inhibits the PTP of a substrate of M(r) approximately 76,000. These results suggest that tyrosine phosphorylation may be involved in transmembrane signalling that mediates platelet adhesion and aggregation in response to pathological shear stresses generated at sites of arterial vaso-occlusion.

Blood ◽  
1987 ◽  
Vol 69 (2) ◽  
pp. 625-628 ◽  
Author(s):  
DM Peterson ◽  
NA Stathopoulos ◽  
TD Giorgio ◽  
JD Hellums ◽  
JL Moake

Different types of platelets in various types of plasma were subjected to levels of shear stress that produce irreversible platelet aggregation in normal platelet-rich plasma (PRP). At shear stresses of 90 or 180 dyne/cm2 applied for 30 seconds or five minutes, aggregation was either absent or only transient and reversible using severe von Willebrand's disease (vWD) PRP (less than 1% von Willebrand factor, vWF); Bernard-Soulier syndrome (BSS) PRP (platelets deficient in the membrane glycoprotein Ib, GPIb); normal PRP plus monoclonal antibody (MoAb) to GPIb; thrombasthenic PRP (platelets deficient in membrane glycoprotein IIb-IIIa complex, GPIIb-IIIa); and normal PRP plus MoAb to GPIIb-IIIa. Shear-induced aggregation was inhibited under the above conditions, even though the platelets were activated to release their granular contents. Sheared normal platelets in vWD plasma aggregated in response to added vWF. These studies demonstrate that the formation of stable platelet aggregates under conditions of high shear requires vWF and the availability of both GPIb and GPIIb-IIIa on platelet membranes. The experiments demonstrate that vWF-platelet interactions can occur in the absence of artificial agonists or chemical modification of vWF. They suggest a possible mechanism for platelet aggregation in stenosed or partially obstructed arterial vessels in which the platelets are subjected to relatively high levels of shear stress.


Blood ◽  
1987 ◽  
Vol 69 (2) ◽  
pp. 625-628 ◽  
Author(s):  
DM Peterson ◽  
NA Stathopoulos ◽  
TD Giorgio ◽  
JD Hellums ◽  
JL Moake

Abstract Different types of platelets in various types of plasma were subjected to levels of shear stress that produce irreversible platelet aggregation in normal platelet-rich plasma (PRP). At shear stresses of 90 or 180 dyne/cm2 applied for 30 seconds or five minutes, aggregation was either absent or only transient and reversible using severe von Willebrand's disease (vWD) PRP (less than 1% von Willebrand factor, vWF); Bernard-Soulier syndrome (BSS) PRP (platelets deficient in the membrane glycoprotein Ib, GPIb); normal PRP plus monoclonal antibody (MoAb) to GPIb; thrombasthenic PRP (platelets deficient in membrane glycoprotein IIb-IIIa complex, GPIIb-IIIa); and normal PRP plus MoAb to GPIIb-IIIa. Shear-induced aggregation was inhibited under the above conditions, even though the platelets were activated to release their granular contents. Sheared normal platelets in vWD plasma aggregated in response to added vWF. These studies demonstrate that the formation of stable platelet aggregates under conditions of high shear requires vWF and the availability of both GPIb and GPIIb-IIIa on platelet membranes. The experiments demonstrate that vWF-platelet interactions can occur in the absence of artificial agonists or chemical modification of vWF. They suggest a possible mechanism for platelet aggregation in stenosed or partially obstructed arterial vessels in which the platelets are subjected to relatively high levels of shear stress.


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.


Blood ◽  
1998 ◽  
Vol 91 (10) ◽  
pp. 3792-3799 ◽  
Author(s):  
Hilde Depraetere ◽  
Nadine Ajzenberg ◽  
Jean-Pierre Girma ◽  
Catherine Lacombe ◽  
Dominique Meyer ◽  
...  

Shear-induced platelet aggregation (SIPA) involves von Willebrand Factor (vWF) binding to platelet glycoprotein (GP)Ib at high shear stress, followed by the activation of αIIbβ3. The purpose of this study was to determine the vWF sequences involved in SIPA by using monoclonal antibodies (MoAbs) to vWF known to interfere with its binding to GPIb and to αIIbβ3. Washed platelets were exposed to shear rates between 100 and 4,000 seconds−1 in a rotational viscometer. SIPA was quantitated by flow cytometry as the disappearance of single platelets (DSP) in the sheared sample in the presence of vWF, relative to a control in the absence of shear and vWF. At a shear rate of 4,000 seconds−1, DSP was increased from 5.9% ± 3.5% in the absence of vWF to 32.7% ± 6.3% in the presence of vWF. This increase in SIPA was not associated with an elevation of P-selectin expression. vWF-dependent SIPA was completely abolished by MoAb 6D1 to GPIb and partially inhibited by MoAb 10E5 to αIIbβ3. Three MoAbs to vWF were compared for their effect on SIPA at 4,000 seconds−1 in the presence of vWF: MoAb 328, known to block vWF binding to GPIb in the presence of ristocetin, MoAb 724 blocking vWF binding to GPIb in the presence of botrocetin, and MoAb 9, an inhibitor of vWF binding to αIIbβ3. Similar to the effect of MoAb 6D1, MoAb 328 completely inhibited the effect of vWF, whereas MoAb 9 had a partial inhibitory effect, as MoAb 10E5 did. In contrast, MoAb 724, as well as its F(ab′)2 fragments, promoted shear-dependent platelet aggregation (165% of the DSP value obtained in the absence of MoAb 724), indicating that MoAb 724 was responsible for an enhanced aggregation, which was independent of binding to the platelet Fcγ receptor. In addition, the enhancement of aggregation induced by MoAb 724 was abrogated by MoAb 6D1 or 10E5 to the level of SIPA obtained in the presence of vWF incubated with a control MoAb to vWF. Finally, the activating effect of MoAb 724 was also found under static conditions at ristocetin concentrations too low to induce platelet aggregation. Our results suggested that on binding to a botrocetin-binding site on vWF, MoAb 724 mimics the effect of botrocetin by inducing an active conformation of vWF that is more sensitive to shear stress or to low ristocetin concentration.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2112-2112
Author(s):  
Xiaoyun Fu ◽  
Ryan P. Gallagher ◽  
Dominic Chung ◽  
Junmei Chen ◽  
José A. López

Abstract Abstract 2112 The interaction between von Willebrand factor (VWF) and the platelet glycoprotein Ib-IX-V complex mediates the first step of platelet adhesion to the vessel wall at sites of injury in the hemostatic response to blood loss. This interaction is also involved in pathologic thrombosis, the most extreme case being thrombotic thrombocytopenic purpura, but the interaction has been proposed to have important pathogenic roles in disparate syndromes such as sepsis, HELLP syndrome, antiphospholipid syndrome, acute lung injury, sickle cell anemia, and cerebral malaria. These syndromes have in common an association with severe inflammation, one of the consequences of which is production of oxidants, in particular by neutrophils. We recently showed that one of the most potent neutrophil oxidants, hypochlorous acid (HOCl), which is produced by the myeloperoxidase-catalyzed reaction of H2O2 with chloride ion, markedly reduces ADAMTS13 proteolysis of VWF by oxidizing M1606 at the ADAMTS13 cleavage site within the A2 domain of VWF (Blood, 115(3) 706-12, 2010). In that study, M1606 present in a substrate A2 peptide was readily oxidized by HOCl, but only minimally oxidized in multimeric plasma VWF, except in the presence of the denaturing agent urea. As this requirement resembled the requirement of urea for ADAMTS13 proteolysis of plasma VWF, we wondered whether the application of shear stress would similarly enhance M1606 oxidation by HOCl. Using a system containing 25 nM MPO (a plasma concentration often seem in inflammatory conditions) and varying concentrations of H2O2, we found that application of 0.6 dynes/cm2 shear stress through a closed circuit of plastic tubing rendered M1606 much more sensitive to oxidation: 80% oxidized within 1 hr. This suggestion of shear-induced unfolding and enhanced oxidation was verified when we examined 7 other methionine residues in the A1A2A3 region of VWF, the region containing the binding sites for platelets and collagen and the ADAMTS13 cleavage site. The Met residues were variably sensitive to oxidation, but all became increasingly oxidized over time in the presence of shear stress. Although the shear stresses we used in this experiment are far below the shear stress considered necessary to unfold even very large VWF multimers, the VWF solution also experienced constant elongational flow generated by a peristaltic pump, necessitating flow acceleration through the region narrowed by the rollers. Elongational flow can impart up to 100-fold more tensile stress to suspended VWF than the constant shear stress (Biophys. J., 98 L35, 2010). Two other findings favor the interpretation that oxidation of the A1A2A3 region is facilitated by domain unfolding. First, we further separated the oxidized VWF by gel-filtration into large, intermediate, and small multimeric fractions and found that methionine oxidation was much more prevalent in the fraction with the largest multimers and rare in the fraction with the smallest multimers. Second, we found that ristocetin, a VWF modulator that simulates the effect of shear stress on VWF, also accelerated oxidation of M1606. In functional tests, we found that HOCl-oxidized plasma VWF agglutinated fixed platelets at concentrations of ristocetin that induced minimal agglutination using unoxidized VWF. These findings have several important clinical implications. First, inflammatory conditions will not only activate endothelial cells and induce release of VWF, especially the largest and most adhesive forms (ultralarge VWF), the oxidants produced from endothelial cells themselves and from the neutrophil respiratory burst will render the VWF resistant to proteolysis. Second, these same oxidants will also convert the largest preexisting plasma VWF multimers that were previously rendered quiescent by ADAMTS13, into hyperfunctional and uncleavable forms. All of these mechanisms converge to generate a highly prothrombotic state, perhaps initially evolved as a mechanism to trap and isolate microorganisms, but which also has the potential to cause tremendous harm to those affected by these inflammatory conditions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2003 ◽  
Vol 102 (6) ◽  
pp. 2122-2129 ◽  
Author(s):  
Shuju Feng ◽  
Julio C. Reséndiz ◽  
Xin Lu ◽  
Michael H. Kroll

Abstract We examined the hypothesis that filamin A binding to the cytoplasmic tail of platelet glycoprotein Ibα (GpIbα) is regulated by pathologic shear stress and modulates von Willebrand factor (VWF)–induced platelet activation. To begin, we examined filamin binding to GpIbα in Chinese hamster ovary cells coexpressing mutant human GpIb-IX and wild-type human filamin A. We observed that many different deletions and truncations N-terminal to GpIbα's cytoplasmic domain residue 594 disrupted filamin A binding, but that binding was unaffected by 14 different point mutations in hydrophilic residues between amino acids 557 and 593. To try to narrow GpIbα's filamin A–binding domain, we next measured the effect of several cytoplasmic domain peptides on human filamin A binding to a GST-GpIbα cytoplasmic domain fusion protein. One peptide (residues 557-575; designated “A4 peptide”) inhibited filamin A binding to the GST-GpIbα cytoplasmic domain fusion protein and competed with GpIbα for binding to filamin A. When the A4 peptide was delivered to intact human platelets using a carrier peptide, we observed the dose-dependent inhibition of VWF-induced platelet aggregation in response to both ristocetin and shear stress. The effect of the A4 peptide on shear-induced platelet aggregation was accompanied by the attenuation of shear-induced filamin A binding to GpIbα and diminished shear-dependent protein tyrosine phosphorylation. These results suggest that shear-dependent VWF-induced platelet activation affects filamin A binding to GpIb-IX-V, and that filamin A binding to the cytoplasmic tail of GpIbα regulates proaggregatory tyrosine kinase signaling.


Blood ◽  
1998 ◽  
Vol 91 (10) ◽  
pp. 3792-3799 ◽  
Author(s):  
Hilde Depraetere ◽  
Nadine Ajzenberg ◽  
Jean-Pierre Girma ◽  
Catherine Lacombe ◽  
Dominique Meyer ◽  
...  

AbstractShear-induced platelet aggregation (SIPA) involves von Willebrand Factor (vWF) binding to platelet glycoprotein (GP)Ib at high shear stress, followed by the activation of αIIbβ3. The purpose of this study was to determine the vWF sequences involved in SIPA by using monoclonal antibodies (MoAbs) to vWF known to interfere with its binding to GPIb and to αIIbβ3. Washed platelets were exposed to shear rates between 100 and 4,000 seconds−1 in a rotational viscometer. SIPA was quantitated by flow cytometry as the disappearance of single platelets (DSP) in the sheared sample in the presence of vWF, relative to a control in the absence of shear and vWF. At a shear rate of 4,000 seconds−1, DSP was increased from 5.9% ± 3.5% in the absence of vWF to 32.7% ± 6.3% in the presence of vWF. This increase in SIPA was not associated with an elevation of P-selectin expression. vWF-dependent SIPA was completely abolished by MoAb 6D1 to GPIb and partially inhibited by MoAb 10E5 to αIIbβ3. Three MoAbs to vWF were compared for their effect on SIPA at 4,000 seconds−1 in the presence of vWF: MoAb 328, known to block vWF binding to GPIb in the presence of ristocetin, MoAb 724 blocking vWF binding to GPIb in the presence of botrocetin, and MoAb 9, an inhibitor of vWF binding to αIIbβ3. Similar to the effect of MoAb 6D1, MoAb 328 completely inhibited the effect of vWF, whereas MoAb 9 had a partial inhibitory effect, as MoAb 10E5 did. In contrast, MoAb 724, as well as its F(ab′)2 fragments, promoted shear-dependent platelet aggregation (165% of the DSP value obtained in the absence of MoAb 724), indicating that MoAb 724 was responsible for an enhanced aggregation, which was independent of binding to the platelet Fcγ receptor. In addition, the enhancement of aggregation induced by MoAb 724 was abrogated by MoAb 6D1 or 10E5 to the level of SIPA obtained in the presence of vWF incubated with a control MoAb to vWF. Finally, the activating effect of MoAb 724 was also found under static conditions at ristocetin concentrations too low to induce platelet aggregation. Our results suggested that on binding to a botrocetin-binding site on vWF, MoAb 724 mimics the effect of botrocetin by inducing an active conformation of vWF that is more sensitive to shear stress or to low ristocetin concentration.


mBio ◽  
2019 ◽  
Vol 10 (2) ◽  
Author(s):  
Felipe Viela ◽  
Valeria Prystopiuk ◽  
Audrey Leprince ◽  
Jacques Mahillon ◽  
Pietro Speziale ◽  
...  

ABSTRACTBinding ofStaphylococcus aureusto the large plasma glycoprotein von Willebrand factor (vWF) is controlled by hydrodynamic flow conditions. Currently, we know little about the molecular details of this shear-stress-dependent interaction. Using single-molecule atomic force microscopy, we demonstrate that vWF binds to theS. aureussurface protein A (SpA) via a previously undescribed force-sensitive mechanism. We identify an extremely strong SpA-vWF interaction, capable of withstanding forces of ∼2 nN, both in laboratory and in clinically relevant methicillin-resistantS. aureus(MRSA) strains. Strong bonds are activated by mechanical stress, consistent with flow experiments revealing that bacteria adhere in larger amounts to vWF surfaces when the shear rate is increased. We suggest that force-enhanced adhesion may involve conformational changes in vWF. Under force, elongation of vWF may lead to the exposure of a high-affinity cryptic SpA-binding site to which bacteria firmly attach. In addition, force-induced structural changes in the SpA domains may also promote strong, high-affinity binding. This force-regulated interaction might be of medical importance as it may play a role in bacterial adherence to platelets and to damaged blood vessels.IMPORTANCEStaphylococcus aureusprotein A (SpA) binds to von Willebrand factor (vWF) under flow. While vWF binding to SpA plays a role inS. aureusadherence to platelets and endothelial cells under shear stress, the molecular basis of this stress-dependent interaction has not yet been elucidated. Here we show that the SpA-vWF interaction is regulated by a new force-dependent mechanism. The results suggest that mechanical extension of vWF may lead to the exposure of a high-affinity cryptic SpA-binding site, consistent with the shear force-controlled functions of vWF. Moreover, strong binding may be promoted by force-induced structural changes in the SpA domains. This study highlights the role of mechanoregulation in controlling the adhesion ofS. aureusand shows promise for the design of small inhibitors capable of blocking colonization under high shear stress.


1991 ◽  
Vol 87 (4) ◽  
pp. 1234-1240 ◽  
Author(s):  
Y Ikeda ◽  
M Handa ◽  
K Kawano ◽  
T Kamata ◽  
M Murata ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document