scholarly journals Shear-Dependent Fibrillogenesis of Fibronectin: Impact of Platelet Integrins and Actin Cytoskeleton

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1443-1443
Author(s):  
Huong T. T. Nguyen ◽  
Khon C. Huynh ◽  
Volker R. Stoldt ◽  
Rudiger E. Scharf

Abstract Introduction: Soluble plasma fibronectin (Fn) with its inactive compact structure requires unfolding to assemble into active fibrils. Fibril formation of Fn is cell-mediated (Mao and Schwarzbauer, 2005) and depending on interactions of Fn with integrin receptors, through binding to αIIbβ3, α5β1, or αvβ3. Less is known about the contribution of biomechanical forces on the fibrillogenesis of Fn. Tension forces generated by cells via cytoskeleton could modulate Fn fibrillogenesis. The aim of this study was to investigate conformational changes of Fn, as induced by (1) platelet integrins, (2) cytoskeletal forces and/or (3) shear rates simulating venous or arterial flow conditions. Methods: Human plasma Fn (100 μg/ml) was added to plates pre-coated with 100 μg/ml of Fn or collagen in the presence or absence of washed platelets (2.5 x 107/ml). Subsequently, the solutions were exposed to shear using a cone-plate rheometer (Haake Rheostress 1). For microscopic analysis (LSM 510, Carl Zeiss), Alexa flour 488-conjugated Fn was used. In parallel experiments, a N-terminal 70kDa fragment of Fn (70 μg/ml) was incubated with soluble Fn at room temperature for 20 min before exposure to shear. To examine the role of distinct platelet integrins on fibril formation of Fn, washed platelets were incubated with monoclonal antibodies LM609, P1D6, 10E5, or abciximab (10 μg/ml, each) for 30 min at room temperature to block αvβ3, α5β1, αIIbβ3, or both αIIbβ3 and αvβ3, prior to the addition of Fn (100 μg/ml) and subsequent exposure to shear. In parallel experiments, washed platelets were pre-incubated with actin-modifying reagents, jasplakinolide (1 μM) or cytochalasin D (10 μM). In all experiments, flow conditions were simulated by shear rates, stepwise increasing from 50 s-1 to 5000 s-1 within 5 min and subsequently decreasing from 5000 s-1 to 50 s-1 within 5 min. Viscosities (mPa's) of shear-exposed solutions were recorded over 10 min. To study the structure of Fn fibrils, solutions were examined by laser scanning microscopy after exposure to shear. To quantify the amount of fibril formation, deoxycholate solubility assays and densitometric analysis of Western blots were performed. Control experiments were conducted under static conditions. Results: Microscopic analyses showed that exposing Fn solutions to shear resulted in fibril formation. Fn fibril diameter varied from 0.5 to 5 μm. Observed fibrils were linked with each other and varied in length (from 50 to 300 μm). Addition of washed platelets to Fn solution resulted in a higher intertwined matrix of fibrils. Treatment of Fn with the N-terminal 70 kDa fragment of Fn, which is known to inhibit Fn matrix assembly, blocked fibril formation of Fn. Western blotting and densitometric analyses revealed that, in the absence of washed platelets, fibril formation (calculated as the ratio of insoluble to soluble Fn) on plates coated with collagen were 2-fold higher than on Fn-immobilized plates (n = 4, p < 0.05). Addition of washed platelets to Fn solution (100 μg/ml) resulted in increases of 20- or 7-fold in fibril formation of Fn, generated by shear on Fn- and collagen-immobilized plates, respectively (p < 0.05, n = 3). In contrast, 10E5 or abxicimab blocking αIIbβ3, or both αIIbβ3 and αvβ3 caused a reduction by 82% or 74% in fibril formation of Fn (p < 0.05, n = 3 each), in comparison to samples without antibodies. Blocking α5β1 or αvβ3by P1D6 or LM609 only caused a reduction by 17% or 56% (p > 0.05, n = 3 each). Incubation of platelets with jasplakinolide, which stabilizes actin, caused an increase in fibril formation by 41%, as compared to samples with untreated platelets (p > 0.05, n = 3). In contrast, disruption of actin by cytochalasin D resulted in a decrease by 86% (p < 0.05, n = 3). Under static conditions, no fibril formation was detected. Conclusions: Our results indicate that fibrillogenesis of Fn is modulated by shear conditions in a surface-dependent manner. Furthermore, formation of fibrils is induced by platelet integrins and actin cytoskeleton. Hereby, αIIbβ3 plays a predominant role, while α5β1 has a minor part among the three examined platelet integrins in terms of Fn fibril formation. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
1997 ◽  
Vol 89 (1) ◽  
pp. 166-175 ◽  
Author(s):  
P.H.M. Kuijper ◽  
H.I. Gallardo Torres ◽  
J.-W.J. Lammers ◽  
J.J. Sixma ◽  
L. Koenderman ◽  
...  

Abstract At sites of vessel wall damage, the primary hemostatic reaction involves platelet and fibrin deposition. At these sites, circulating leukocytes marginate and become activated. Adhered platelets can support leukocyte localization; however, the role of fibrin in this respect is not known. We studied the adhesion of human neutrophils (polymorphonuclear leukocytes [PMNs]) to endothelial extracellular matrix (ECM)-bound fibrin and platelets under flow conditions. ECM alone did not show PMN adhesion. ECM-coated cover slips were perfused with plasma to form a surface-bound fibrin network, and/or with whole blood to allow platelet adhesion. Unstimulated PMNs adhered to fibrin at moderate shear stress (20 to 200 mPa). ECM-bound platelets induced rolling adhesion and allowed more PMNs to adhere at higher shear (320 mPa). ECM coated with both platelets and fibrin induced more static and shear-resistant PMN adhesion. PMN adhesion to fibrin alone but not to platelet/fibrin surfaces was inhibited by soluble fibrinogen. Adhesion to fibrin alone was inhibited by CD11b and CD18 blocking antibodies. Furthermore, fibrin formed under flow conditions showed up to threefold higher PMN adhesion compared with fibrin formed under static conditions, due to structural differences. These results indicate that circulating PMNs adhere to fibrin in an integrin-dependent manner at moderate shear stresses. However, at higher shear rates (<200 mPa), additional mechanisms (ie, activated platelets) are necessary for an interaction of PMNs with a fibrin network.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3519-3519
Author(s):  
Huong T. T. Nguyen ◽  
Khon C. Huynh ◽  
Volker R. Stoldt ◽  
Rüdiger E. Scharf

Abstract Introduction Soluble plasma fibronectin (Fn) with its inactive compact structure needs to be unfolded to assemble into active fibrils. Fibril formation of Fn is cell-mediated and depends on interactions between Fn and integrin receptors, through binding to αIIbβ3, α5β1, or αvβ3. Less is known about the contribution of biomechanical forces on the fibrillogenesis of Fn. Mechanical forces assessed by atomic force microscopy were shown to regulate the transformation of Fn from its compact structure to its extended fibrillar state when cryptic binding sites of Fn type III repeats were substituted (Gao et al., 2003). The aim of this study was to investigate conformational changes of Fn, as induced (1) by platelet integrin receptors and/or (2) by shear rates simulating venous or arterial flow conditions. Methods Fn isolated from fresh frozen human plasma was added, at different concentrations (50 or 100 μg/ml), to plates pre-coated with 100 μg/ml of soluble Fn or BSA. Subsequently, the solutions were exposed to shear generated by different stainless steel cones with bare surfaces, which were used as controls, or which were pre-grafted with O,O’-Bis(amino-propyl) polyethylene glycol (PEG) once or twice to reduce non-specific adsorption of plasma proteins. To study the effects of platelet on conformational changes of Fn, in the absence or presence of washed platelets (2.5 x 107/ml or 2.5 x 106/ml), soluble Fn (100 μg/ml) was exposed to shear generated by cones grafted with PEG twice. To examine the role of distinct platelet integrins on fibril formation of Fn, washed platelets (2.5 x 107/ml) were incubated with the monoclonal antibodies LM609, P1D6, 10E5, or abciximab (10 μg/ml, each) for 30 min at room temperature to block αvβ3, α5β1, αIIbβ3, or both αIIbβ3 and αvβ3, respectively, prior to the addition of Fn (100 μg/ml) and shear exposure. In all experiments, flow conditions were simulated by dynamic shear rates stepwise increasing from 50 s-1 to 5000 s-1 within 5 min and subsequently decreasing from 5000 s-1 to 50 s-1 within 5 min using a cone-plate rheometer (Haake Rheostress 1). Viscosities (mPa s) of shear-exposed solutions were recorded over 10 min. To quantify the amount of fibril formation, DOC solubility assays and Western blotting were performed. Control experiments were conducted under static conditions. Results Upon exposure to shear stress, the viscosity in the sample increased, suggesting conformational changes in Fn. Western blotting and densitometric analyses revealed that, under shear generated by untreated cones, the ratios of insoluble to soluble Fn (indicative of fibril formation) increased significantly from 0.018 ± 0.012 (mean ratio ± SD) to 0.121 ± 0.08 (p < 0.05, n = 4) (Fn-immobilized plates) and from 0.021 ± 0.009 to 0.059 ± 0.022 (p < 0.05, n = 4) (BSA-immobilized plates) when the concentration of added soluble Fn was elevated from 50 μg/ml to 100 μg/ml. The observed fibril formation of Fn was significantly lower when using cones grafted with PEG once or twice, in comparison to bare steel cones (p < 0.05, n = 4). Addition of washed platelets to Fn solution (100 μg/ml) resulted in significant increases of 8- and 20-fold in fibril formation of Fn, generated by shear on BSA- and Fn-immobilized plates, respectively (p < 0.05, n = 3). In contrast, using 10E5 or abxicimab to block αIIbβ3, or both αIIbβ3 and αvβ3 caused a reduction by 82% or 74% in fibril formation of Fn, respectively (n = 3), in comparison to samples without antibodies. Blocking α5β1 or αvβ3 by P1D6 or LM609 only caused a reduction by 17% or 56%, respectively (n = 3). Under static conditions, no fibril formation was detected. Conclusions Our results indicate that the fibril formation of Fn solution under shear can be monitored by changes in its viscosity. In addition, fibrillogenesis of Fn is modulated by shear conditions and physical properties of stainless steel. Furthermore, the formation of fibrils depends on the Fn concentration and is modulated by platelet integrins. Hereby, αIIbβ3 plays a predominant role, while α5β1 has a minor part among the three examined platelet integrins, in terms of fibril formation. Our system provides useful information regarding (1) surface- and shear-induced alterations of unfolding of Fn and (2) the contribution of its binding partners, including β3-integrins and α5β1. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1997 ◽  
Vol 89 (1) ◽  
pp. 166-175 ◽  
Author(s):  
P.H.M. Kuijper ◽  
H.I. Gallardo Torres ◽  
J.-W.J. Lammers ◽  
J.J. Sixma ◽  
L. Koenderman ◽  
...  

At sites of vessel wall damage, the primary hemostatic reaction involves platelet and fibrin deposition. At these sites, circulating leukocytes marginate and become activated. Adhered platelets can support leukocyte localization; however, the role of fibrin in this respect is not known. We studied the adhesion of human neutrophils (polymorphonuclear leukocytes [PMNs]) to endothelial extracellular matrix (ECM)-bound fibrin and platelets under flow conditions. ECM alone did not show PMN adhesion. ECM-coated cover slips were perfused with plasma to form a surface-bound fibrin network, and/or with whole blood to allow platelet adhesion. Unstimulated PMNs adhered to fibrin at moderate shear stress (20 to 200 mPa). ECM-bound platelets induced rolling adhesion and allowed more PMNs to adhere at higher shear (320 mPa). ECM coated with both platelets and fibrin induced more static and shear-resistant PMN adhesion. PMN adhesion to fibrin alone but not to platelet/fibrin surfaces was inhibited by soluble fibrinogen. Adhesion to fibrin alone was inhibited by CD11b and CD18 blocking antibodies. Furthermore, fibrin formed under flow conditions showed up to threefold higher PMN adhesion compared with fibrin formed under static conditions, due to structural differences. These results indicate that circulating PMNs adhere to fibrin in an integrin-dependent manner at moderate shear stresses. However, at higher shear rates (<200 mPa), additional mechanisms (ie, activated platelets) are necessary for an interaction of PMNs with a fibrin network.


2020 ◽  
Vol 21 (22) ◽  
pp. 8526
Author(s):  
Angela Di Somma ◽  
Federica Recupido ◽  
Arianna Cirillo ◽  
Alessia Romano ◽  
Alessandra Romanelli ◽  
...  

Biofilms consist of a complex microbial community adhering to biotic or abiotic surfaces and enclosed within a protein/polysaccharide self-produced matrix. The formation of this structure represents the most important adaptive mechanism that leads to antibacterial resistance, and therefore, closely connected to pathogenicity. Antimicrobial peptides (AMPs) could represent attractive candidates for the design of new antibiotics because of their specific characteristics. AMPs show a broad activity spectrum, a relative selectivity towards their targets (microbial membranes), the ability to act on both proliferative and quiescent cells, a rapid mechanism of action, and above all, a low propensity for developing resistance. This article investigates the effect at subMIC concentrations of Temporin-L (TL) on biofilm formation in Pseudomonas fluorescens (P. fluorescens) both in static and dynamic conditions, showing that TL displays antibiofilm properties. Biofilm formation in static conditions was analyzed by the Crystal Violet assay. Investigation of biofilms in dynamic conditions was performed in a commercial microfluidic device consisting of a microflow chamber to simulate real flow conditions in the human body. Biofilm morphology was examined using Confocal Laser Scanning Microscopy and quantified via image analysis. The investigation of TL effects on P. fluorescens showed that when subMIC concentrations of this peptide were added during bacterial growth, TL exerted antibiofilm activity, impairing biofilm formation both in static and dynamic conditions. Moreover, TL also affects mature biofilm as confocal microscopy analyses showed that a large portion of preformed biofilm architecture was clearly perturbed by the peptide addition with a significative decrease of all the biofilm surface properties and the overall biomass. Finally, in these conditions, TL did not affect bacterial cells as the live/dead cell ratio remained unchanged without any increase in damaged cells, confirming an actual antibiofilm activity of the peptide.


2000 ◽  
Vol 347 (1) ◽  
pp. 183-192 ◽  
Author(s):  
Juan A. ROSADO ◽  
Stewart O. SAGE

We have investigated the mechanism of Ca2+ entry into fura-2-loaded human platelets by preventing the prenylation of proteins such as small GTP-binding proteins. The farnesylcysteine analogues farnesylthioacetic acid (FTA) and N-acetyl-S-geranylgeranyl-L-cysteine (AGGC), which are inhibitors of the methylation of prenylated and geranylgeranylated proteins respectively, significantly decreased thrombin-evoked increases in intracellular free Ca2+ concentration ([Ca2+]i) in the presence, but not in the absence, of external Ca2+, suggesting a relatively selective inhibition of Ca2+ entry over internal release. Both these compounds and N-acetyl-S-farnesyl-L-cysteine, which had similar effects to those of FTA, also decreased Ca2+ entry evoked by the depletion of intracellular Ca2+ stores with thapsigargin. The inactive control N-acetyl-S-geranyl-L-cysteine was without effect. Patulin, an inhibitor of prenylation that is inert with respect to methyltransferases, also decreased store-regulated Ca2+ entry. Cytochalasin D, an inhibitor of actin polymerization, significantly decreased store-regulated Ca2+ entry in a time-dependent manner. Both cytochalasin D and the farnesylcysteine analogues FTA and AGGC inhibited actin polymerization; however, when evoking the same extent of decrease in actin filament formation, FTA and AGGC showed greater inhibitory effects on Ca2+ entry, indicating a cytoskeleton-independent component in the regulation of Ca2+ entry by small GTP-binding-protein. These findings suggest that prenylated proteins such as small GTP-binding proteins are involved in store-regulated Ca2+ entry through actin cytoskeleton-dependent and cytoskeleton-independent mechanisms in human platelets.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 5191-5191
Author(s):  
Khon C. Huynh ◽  
Volker R. Stoldt ◽  
Marianna Gyenes ◽  
Rüdiger E. Scharf

Abstract Abstract 5191 Introduction: To fulfill their role in hemostasis, circulating platelets need to irreversibly adhere to the site of vascular injury and to resist to shear stress generated by the flowing blood. We previously reported that there is a relationship between the conformation of fibronectin (Fn) and its role in platelet adhesion and aggregation (Huynh, K. C. et al., ASH Annual Meeting Abstract, 2011. 118(21): p. 2209). In the present study, we examined the effect of shear stress on the assembly of Fn by adherent platelets. Moreover, we studied the role of β3 integrins (αIIbβ3 and αvβ3) in Fn assembly under flow conditions. Methods: Alexa fluor 488-conjugated fibronetin (Fn488) was added to suspensions of washed platelets (108/ml) in HEPES Tyrode buffer. CaCl2 (2 mM) and ADP (10 μM) were added immediately prior to the experiments. The samples (150 μl) were subsequently applied onto plates precoated with 50 μg/ml Fn. A DiaMed Impact-R device was used to generate shear rates of 500 s−1 or 5000 s−1 for 2 min or 10 min. Nonadherent platelets were removed by washing with PBS buffer followed by addition of 150 μl of 2 % DOC lysis buffer. Lysates were collected and total protein concentrations were determined by Bradford assay. The DOC-insoluble pellets containing Fn fibrils were isolated by centrifugation at 13, 500 rpm for 20 min. Pellets were then solubilized with 100 μl of 1 % SDS buffer. Equal amounts of samples based on total protein concentrations were loaded onto wells of 96-well microplates. Fluorescence signals from Fn488 of samples were recorded by a Fluoroskan microplate reader. In some experiments, abciximab (anti-β3, 10 μg/ml) or LM609 (anti-αvβ3, 5 μg/ml) antibody, were added to platelet mixtures before loading onto Fn precoated plates. All data were collected from at least three different experiments and analyzed using GraphPad Quickcals. To test for statistical differences, student's t-test was used. Results: Fn assembly by adherent platelets was strongly affected by the applied shear rate but not by the exposure time to shear. At a shear rate of 500 s−1, there were no insoluble Fn fibrils detectable in samples with adherent platelets after 2 or 10 min. When shear rates increased from 500 s−1 to 5000 s−1, the amount of insoluble Fn detectable on platelets after 2 and 10 min increased significantly (p < 0. 05) suggesting that adherent platelets exposed to high shear rates assemble more Fn fibrils on their surface. However, prolongation of exposure time to shear from 2 to 10 min did not result in significantly more Fn assembled by adherent platelets. By contrast, there were no insoluble fibrils that could be detected with adherent platelets under static conditions for 2 and 10 min. After 2 min at a shear rate of 5000 s−1, platelets blocked with abciximab showed a significant decrease in the amounts of insoluble Fn fibrils in comparison with control experiments (no antibody) (p = 0. 02). Similar inhibitory effects could be seen with platelets treated with LM609. In parallel experiments in which 10 min at 5000 s−1 were applied, both abciximab and LM609 had an inhibitory effect on Fn fibrillogenesis with a stronger effect by abciximab. Taken together, these data show that αvβ3 even at the low expression on platelets plays a major role in initiating the fibrillogenesis of Fn under high shear rate conditions, whereas αIIbβ3 contributes to the progression of Fn fibrils formation subsequently. Conclusion: Our observations document that the assembly of Fn on the surface of adherent platelets is strongly affected by shear rate conditions. In addition, our data imply that, despite its lower expression on platelet surface, αvβ3 provides a significant contribution in initiating the Fn assembly under high flow conditions, as compared with αIIbβ3. By contrast, αIIbβ3 with its abundant amount on the platelet surface probably exerts its effect in the later phase of Fn fibrillogenesis. The present findings support the contention that not a single integrin or Fn binding domain, but multiple interaction steps including different molecules and Fn domains may be involved in assembling Fn. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 2013 ◽  
pp. 1-5
Author(s):  
Dang-liang Wang ◽  
Han-ying Bai ◽  
Gao Yue

Urea-formaldehyde resin (UFR), one of chemical grouts in which the major ingredients are urea-formaldehyde and resin, is widely used in Chinese coal mines grouting. The gel characteristics of urea-formaldehyde resin (UFR) chemical grout under static conditions have been studied by many researchers. However, there is little research carried out on the gel characteristics under shear flow conditions. In fact, chemical grout like UFR keeps in shear flow conditions before gelling in the grouting process. In order to investigate the gel characteristics of UFR in shear flow conditions, an apparatus which consists of a magnetic stirrer and a viscometer was established. Magnetic stirrer was used to shear UFR at different velocity. Then the changes of UFR viscosity could be recorded by viscometer. As a result, the gel characteristics were summarized under different shear rates, and a formula of gelling is derived. The results show that the grouting flow rate influences the gelling time. Faster flow rates will cause longer gelling time, which means that the time for the grout to gel during the flowing process under shear flow conditions is longer than that under static conditions.


Blood ◽  
1988 ◽  
Vol 72 (1) ◽  
pp. 82-88
Author(s):  
PF Nievelstein ◽  
JJ Sixma

Previous studies have indicated that activated blood platelets interact with fibronectin through binding of fibronectin to the glycoprotein IIb- IIIa complex (GPIIb-IIIa). The cell attachment site of fibronectin with its crucial arg-gly-asp(-ser) [RGD(S)]sequence is involved in these bindings. We studied the importance of these interactions for the fibronectin dependence of platelet adhesion under flow conditions. An RGDS-containing hexapeptide (GRGDSP) was compared with a nonreactive control peptide (GRGESP). The GRGDSP-peptide inhibited thrombin-induced aggregation and adhesion under static conditions at 0.1 mmol/L. This concentration had no effect on platelet adhesion to nonfibrillar collagen type I in flow. GRGDSP at 1 mmol/L had a significant inhibitory effect at 1,500 s-1, but not at the lower shear rates of 800 and 300 s-1 where platelet adhesion is also fibronectin dependent. On the matrix of cultured human umbilical vein endothelial cells, 1 mmol/L GRGDSP had no effect on platelet adhesion. The relation between GPIIb- IIIa and fibronectin dependence was investigated with platelets of a patient with Glanzmann's thrombasthenia and monoclonal antibodies to GPIIb-IIIa using endothelial cell matrix (ECM) as a surface. Platelets of normal controls or a patient with Glanzmann's thrombasthenia showed a similar inhibition of adhesion in the presence of fibronectin-free plasma after the ECMs had been preincubated with antifibronectin F(ab')2 fragments. Incubation of platelets with anti-GPIIb-IIIa showed inhibition of platelet adhesion at high shear rates. Dependence on fibronectin for platelet adhesion was still observed even though separate experiments had shown that these anti-GPIIb-IIIa antibodies could block binding of radiolabeled fibronectin to thrombin-activated platelets. These data suggest the existence of another binding system for the interaction of platelets with fibronectin that may only appear when fibronectin is present on a surface.


Blood ◽  
1988 ◽  
Vol 72 (1) ◽  
pp. 82-88 ◽  
Author(s):  
PF Nievelstein ◽  
JJ Sixma

Abstract Previous studies have indicated that activated blood platelets interact with fibronectin through binding of fibronectin to the glycoprotein IIb- IIIa complex (GPIIb-IIIa). The cell attachment site of fibronectin with its crucial arg-gly-asp(-ser) [RGD(S)]sequence is involved in these bindings. We studied the importance of these interactions for the fibronectin dependence of platelet adhesion under flow conditions. An RGDS-containing hexapeptide (GRGDSP) was compared with a nonreactive control peptide (GRGESP). The GRGDSP-peptide inhibited thrombin-induced aggregation and adhesion under static conditions at 0.1 mmol/L. This concentration had no effect on platelet adhesion to nonfibrillar collagen type I in flow. GRGDSP at 1 mmol/L had a significant inhibitory effect at 1,500 s-1, but not at the lower shear rates of 800 and 300 s-1 where platelet adhesion is also fibronectin dependent. On the matrix of cultured human umbilical vein endothelial cells, 1 mmol/L GRGDSP had no effect on platelet adhesion. The relation between GPIIb- IIIa and fibronectin dependence was investigated with platelets of a patient with Glanzmann's thrombasthenia and monoclonal antibodies to GPIIb-IIIa using endothelial cell matrix (ECM) as a surface. Platelets of normal controls or a patient with Glanzmann's thrombasthenia showed a similar inhibition of adhesion in the presence of fibronectin-free plasma after the ECMs had been preincubated with antifibronectin F(ab')2 fragments. Incubation of platelets with anti-GPIIb-IIIa showed inhibition of platelet adhesion at high shear rates. Dependence on fibronectin for platelet adhesion was still observed even though separate experiments had shown that these anti-GPIIb-IIIa antibodies could block binding of radiolabeled fibronectin to thrombin-activated platelets. These data suggest the existence of another binding system for the interaction of platelets with fibronectin that may only appear when fibronectin is present on a surface.


2013 ◽  
Vol 109 (01) ◽  
pp. 61-71 ◽  
Author(s):  
Mah Pui Mei ◽  
Yu Pei Xiao ◽  
Anthony Maraveyas ◽  
Camille Ettelaie ◽  
Mary E. W. Collier

SummaryThe role of tissue factor (TF)-containing microparticles in clot propagation has been established, but the ability of circulating microparticles to initiate coagulation has been disputed. However, TF-bearing microparticles, particularly endothelial-microparticles generated during disease, may interact with extracellular matrices which in turn can localise circulating TF to sites of injury. In order to examine this hypothesis in vitro, microparticles were isolated from human coronary artery endothelial cells transfected to overexpress TF, tumour-necrosis factor (TNF)α-treated cells or non-transfected cells lacking TF. The ability of microparticles to bind collagen-IV, fibronectin and fibrin was examined under static conditions and arterial shear rates (650 s-1), and also in the presence of inhibitory antibodies against β1-, β3-, α3-and αv-integrins or an anti-TF antibody. TF-microparticles showed increases of up to 43% and 24% in adherence to collagen-IV and fibronectin, respectively, compared to control microparticles under shear flow. Furthermore, TF-containing microparticles, but not the transfected parent cells had increased levels of β1-integrin compared to TF-deficient microparticles. Pre-incubation of microparticles with a β1-integrin-blocking antibody counteracted the additional adhesion of TFmicroparticles compared to control microparticles. Finally, adherence of TF microparticles to collagen-IV or fibronectin resulted in increased TF activity by concentrating TF onto the surface. In conclusion, the presence of TF within microparticles enhances the interactions of endothelial cell-derived microparticles with extracellular matrices in an integrin-dependent manner. Accumulation and localisation of these microparticles in turn results in the enhancement of TF activity. This may be an innate mechanism by which TF-bearing microparticles induce coagulation upon vascular injury.


Sign in / Sign up

Export Citation Format

Share Document