首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
It has been previously shown that fibrinogen (FG) associates specifically with human umbilical vein and bovine aortic endothelial cells (EC) in culture and induces EC migration. In the present study, we have investigated whether the FG-EC interaction is mediated by an Arg-Gly-Asp (RGD) recognition specificity and whether EC membrane proteins related to platelet GPIIb-IIIa are involved. Highly purified radioiodinated human FG, containing no detectable fibronectin, interacted with cultured human and bovine EC in suspension in a time-dependent and specific manner. The binding was inhibited by EDTA. Two polyclonal antibodies to platelet GPIIb-IIIa, which immunoprecipitated a heterodimer molecule from EC, inhibited FG binding to EC. These same antibodies inhibited FG-induced EC migration in a dose-dependent manner as measured in a Boyden chamber. Preabsorption of the antibodies with purified platelet GPIIb-IIIa markedly reduced both inhibitory activities. A series of synthetic RGD-containing peptides inhibited FG binding to EC and FG-induced EC migration. Gly-Arg-Gly-Asp (GRGD) was the most active peptide tested in inhibiting FG binding and EC migration (ID50 of 30 microM), and conservative substitutions in the RGD sequence markedly reduced inhibitory activity (ID50 greater than 1,000 microM). These results indicate that FG binding and EC migration are events mediated by an RGD recognition specificity and that EC surface proteins immunologically related to the GPIIb-IIIa complex on platelets are involved in this recognition.  相似文献   

2.
Parker  RI; Gralnick  HR 《Blood》1987,70(5):1589-1594
This study demonstrates that when platelets are stimulated by thrombin in the presence of low concentrations of purified human fibrinogen (10 to 20 micrograms/mL, final concentration) binding of released platelet von Willebrand factor (plt-vWF) to the platelet membrane is enhanced. This effect appears to be mediated by fibrin monomer produced by the action of thrombin on the fibrinogen in the incubation suspension. When fibrin polymerization is inhibited, the binding of released plt-vWF to the platelets is markedly increased. This enhanced binding is dependent on platelet glycoprotein Ib (GPIb) as shown by a decreased response with Bernard-Soulier platelets and inhibition by both monoclonal and polyclonal antibodies against glycocalicin. The binding of fibrin to thrombin-activated platelets preincubated with monoclonal antibody against GPIIb/IIIa is increased when the predominant form of fibrin is fibrin monomer. The fibrin binding is also decreased in the presence of antibody against glycocalicin. Our data demonstrate that fibrin monomer facilitates plt-vWF binding to the glycocalicin portion of platelet GPIb on thrombin-stimulated platelets and that binding of fibrin monomer to glycocalicin is necessary for this response to occur.  相似文献   

3.
Thrombospondin promotes platelet aggregation   总被引:11,自引:1,他引:11  
Thrombospondin (TSP), isolated from human platelets, promotes aggregation of both nonstimulated platelets and platelets stimulated with thrombin or ADP. The TSP-promoted aggregation is specific since a monoclonal antibody against TSP inhibits the effect of exogenously added TSP and inhibits thrombin-induced platelet aggregation in the absence of added TSP. Several lines of evidence suggest that TSP mediates its effect on aggregation of nonstimulated and stimulated platelets through different platelet-surface receptor systems. The TSP- promoted aggregation of nonstimulated platelets was inhibited by a monoclonal antibody to platelet glycoprotein IV (GPIV), but not by a monoclonal antibody to the fibrinogen receptor, GPIIb-IIIa. In contrast, the antibody to GPIIb-IIIa totally inhibited the TSP- potentiated aggregation of thrombin-stimulated platelets, whereas the antibody to GPIV has no effect. Thus, these studies suggest that TSP promotes platelet aggregation by at least two mechanisms--one dependent on and one independent of the platelet fibrinogen receptor system.  相似文献   

4.
Naimushin YA  Mazurov AV 《Platelets》2004,15(7):419-425
In this study we investigated mechanisms of platelet interaction with von Willebrand factor (vWF) induced by activating anti-glycoprotein (GP)IIb-IIIa antibody CRC54 directed against LIBS (ligand-induced binding site epitope) in GPIIIa. It was demonstrated that aggregation of washed platelets (measured in Born aggregometer) could be stimulated by CRC54 not only in the presence of fibrinogen but vWF as well. The level of aggregation induced in the presence of saturating concentrations of vWF (approximately 80 microg/ml) was even higher than that in the presence of 1 mg/ml of fibrinogen. Aggregation supported by vWF unlike fibrinogen supported aggregation was almost completely inhibited not only by GPIIb-IIIa antagonists (F(ab')2 fragment of blocking anti-GPIIb-IIIa antibody CRC64 and peptidomimetic aggrastat) but also by anti-GPIb blocking antibody AK2. Aggregation response induced by CRC54 in the presence of vWF was much lower when normal platelets were substituted with GPIb-deficient platelets and this residual aggregation was not affected by anti-GPIb antibody AK2 but still inhibited by anti-GPIIb-IIIa blocking antibody fragment. CRC54-induced aggregation supported by vWF (as well as by fibrinogen) was only partially inhibited by prostaglandin E1, indicating that at least its initiation does not require activation of platelets. CRC54, both in the presence of vWF and fibrinogen, failed to stimulate serotonin secretion at physiological Ca2+ concentration of 1 mM, although substantial release reaction was detected when Ca2+ concentration was decreased to 0.1 mM. CRC54 could also stimulate platelet interaction with immobilized vWF and fibrinogen. However, unlike platelet aggregation in suspension mediated by flow phase vWF, platelet adhesion to adsorbed vWF (in a same way as to fibrinogen) was inhibited only by GPIIb-IIIa but not GPIb antagonists. The data obtained indicated that vWF support platelet aggregation induced by activating anti-GPIIb-IIIa via interaction with two receptors - activated GPIIb-IIIa and GPIb.  相似文献   

5.
Plow  EF; Marguerie  GA; Ginsberg  MH 《Blood》1985,66(1):26-32
Plasma fibronectin binds in a specific and saturable manner to thrombin- stimulated platelets. gamma-Thrombin stimulated 80% as much fibronectin binding to platelets as alpha-thrombin with conversion of less than or equal to 1% of platelet fibrinogen to fibrin. Afibrinogenemic and normal platelets bound similar quantities of fibronectin in the presence of calcium or magnesium-ethylene glycol tetra-acetic acid (EGTA). These observations indicate that fibronectin can interact with platelets without involvement of fibrin or fibrinogen. Nevertheless, two different effects of fibrin(ogen) on fibronectin binding were observed. First, exogenous fibrinogen inhibited fibronectin binding to thrombin-stimulated platelets. This inhibition was unidirectional, as fibronectin did not inhibit fibrinogen binding to ADP or thrombin- stimulated cells. Second, formaldehyde-fixed cells with surface- associated fibrin bound significant quantities of fibronectin. This interaction required calcium and did not occur on fixed cells with or without surface-bound fibrinogen. A portion of the ligand bound to fixed cells with surface-associated fibrin was modified to form a derivative with a molecular weight identical to that of the fibronectin subunit cross-linked to the alpha-chain of fibrin. This high mol wt derivative was also observed to a variable extent with living cells in the presence of magnesium or calcium but not in the presence of magnesium-EGTA. Thus, fibronectin binds to platelets by at least two mechanisms: (1) a fibrin(ogen)-independent pathway that requires divalent ions and is inhibited by exogenous fibrinogen; and (2) a fibrin-dependent pathway with an absolute calcium requirement. With nonaggregated, thrombin-stimulated platelets, the former pathway appears to predominate.  相似文献   

6.
Porcine von Willebrand factor (PvWF) induces platelet aggregation which is thought to be responsible for the thrombocytopenia that occurs in haemophilic patients treated with commercial preparations of porcine factor VIII. This study demonstrates that such aggregation can be completely inhibited by a monoclonal antibody against human platelet glycoprotein GPIb and partially inhibited by an antibody directed against platelet GPIIb/IIIa. The interaction of PvWF with GPIb is also demonstrated by the inhibitory effect of purified glycocalycin on aggregation. The binding site of PvWF to GPIb is very close to that of human vWF, since a recombinant peptide blocks the binding of both molecules to GPIb. When platelets are incubated with PvWF, the GPIIb/IIIa receptor is activated and binds fibrinogen. PvWF also binds to GPIIb/IIIa when platelets are stimulated with thrombin, suggesting that the molecule has the same RGD sequence as other adhesive proteins (human vWF, fibrinogen, fibronectin and vitronectin). These findings identify the dual mechanisms responsible for in vivo platelet aggregation induced by PvWF, i.e. binding to GPIb and activation of the GPIIb/IIIa receptor.  相似文献   

7.
Sloan SM  Brown EB  Liu Q  Frojmovic MM 《Platelets》2000,11(2):99-110
Although platelet cross-bridging is mediated primarily by the binding of fibrinogen to its activated membrane receptor, glycoprotein (GP) IIb-IIIa*, such an interaction may not be sufficient to support the aggregation process. As this question could potentially be answered by reconstituting GPIIb-IIIa* into a non-platelet environment such as liposomes, a protocol was developed for the generation of large lipid vesicles containing purified GPIIb-IIIa*. Flow cytometric techniques confirmed that the receptor was present in the lipid bilayer and were used to evaluate the characteristics of fibrinogen binding to the liposomes, which like fibrinogen-platelet interactions exhibited specificity, saturability, time dependence and calcium dependence. No fibrinogen-specific aggregation of GPIIb-IIIa* liposomes with stir or shear was observed, as determined by flow cytometric cell counting and microscopic examination of particles. In contrast, activated platelets rapidly bound Fg and rapidly formed large aggregates. The Fg associated with GPIIb-IIIa* in liposomes was 'normally' recognized by two fluorescently labelled antibodies: 4A5, which interacts with the Fg gamma chain C-terminus (residues 400-411) required for Fg-mediated cross-bridging of activated platelets in platelet aggregation (Shiba E, Lindon JN, Kushner L, Matsueda GR, Hawiger J, Kloczewiak M, Kudryk B, Salzman EW. Antibody-detectable changes in fibrinogen adsorption affecting platelet activation on polymer surfaces. Am J Physiol 1991; 260: C965-74), and anti-Fg-RIBS-I, which associates with an epitope on Fg (residues 373-385) expressed upon binding to GPIIb-IIIa. These data suggest that the Fg gamma-terminus is sterically accessible for particle cross-bridging and that an identical conformational change occurs for receptor-bound Fg on both liposomes and platelets. It thus appears that cellular elements aside from GPIIb-IIIa, such as cytoskeletal proteins proposed to be necessary for receptor 'anchoring', play a necessary role in flow-associated platelet aggregation.  相似文献   

8.
Mondoro TH  White MM  Jennings LK 《Blood》2000,96(7):2487-2495
Glycoprotein (GP) IIb-IIIa plays a critical role in platelet aggregation and platelet-mediated clot retraction. This study examined the intramolecular relationship between GPIIb-IIIa activation and fibrinogen binding, platelet aggregation, and platelet-mediated clot retraction. To distinguish between different high-affinity activation states of GPIIb-IIIa, the properties of an antibody (D3) specific for GPIIIa that induces GPIIb-IIIa binding to adhesive protein molecules and yet completely inhibits clot retraction were used. Clot retraction inhibition by D3 was not due to altered platelet-fibrin interaction; however, combination treatments of D3 and adenosine diphosphate (ADP) inhibited full-scale aggregation and decreased the amounts of GPIIb-IIIa and talin incorporated into the core cytoskeletons. Morphologic evaluation of the D3/ADP aggregates showed platelets that were activated but to a lesser extent when compared to ADP only. ADP addition to platelets caused an increase in the number of D3 binding sites indicating that ligand had bound to the GPIIb-IIIa receptor. These data suggest that high-affinity GPIIb-IIIa- mediated ligand binding can be separated mechanistically from GPIIb-IIIa-mediated clot retraction and that clot retraction requires additional signaling through GPIIb-IIIa after ligand binding. The conformation recognized by D3 represents the expression of a GPIIb-IIIa activation state that participates in full-scale platelet aggregation, cytoskeletal reorganization, and clot retraction.  相似文献   

9.
Platelet-to-platelet aggregation is critical to the formation of hemostatic thrombi which limit bleeding following vascular injury and also contributes to obstructive thrombi in acute myocardial infarction, stroke, or other thrombotic diseases. Platelet aggregation is mediated by platelet surface glycoprotein (GP) IIb-IIIa (integrin αIIbβ3, CD41/61) on adjacent platelets. Upon platelet activation by adenosine diphosphate (ADP), thrombin, or other platelet agonists, GPIIb-IIIa undergoes conformational changes from a “resting” bent conformation to an “activated” extended conformation. In GPIIb-IIIa’s activated conformation, a binding site is exposed which interacts with the arginine-glycine-aspartic acid (RGD) residues in the fibrinogen alpha chain, permitting fibrinogen binding and cross-bridging of adjacent activated platelets. Consequently, changes in the state of GPIIb-IIIa activation closely correlate with fibrinogen binding and the degree of platelet-platelet aggregation. In contrast to radiolabeled ligand methods used for bulk receptor-binding studies, flow cytometry allows the rapid analysis of fibrinogen receptor expression on single cells, thereby enabling analysis of the kinetics of GPIIb-IIIa activation and differences between platelets in their expression of activated GPIIb-IIIa. The present review will consider the use of flow cytometry to monitor GPIIb-IIIa activation and its application in clinical and research settings.  相似文献   

10.
We have characterized a murine IgG monoclonal antibody, OP-G2, specific for platelet glycoprotein (GP) IIb-IIIa (alpha IIb beta 3). OP-G2 Fab fragments inhibit fibrinogen-mediated platelet aggregation and competitively inhibit adenosine diphosphate-induced binding of 125I-fibrinogen to washed platelets. OP-G2 binding to GPIIb-IIIa is specifically inhibited by RGD-containing peptides but not the fibrinogen gamma-chain carboxy-terminal peptide, and OP-G2 Fab fragments, like RGD-containing peptides, alter the conformation of GPIIb-IIIa resulting in the expression of a ligand-induced binding site (LIBS) recognized by PMI-1. OP-G2 fails to bind to the recombinant Cam variant of GPIIb-IIIa (alpha III beta 3Cam) wherein an Asp119 to Tyr119 substitution in GPIIIa abrogates the ability to recognize RGD. These data indicate that OP-G2 recognizes an epitope at or in very close proximity to the RGD recognition site of GPIIb-IIIa and that, in every aspect tested, OP-G2 behaves like a macromolecular RGD ligand. Interestingly, two-color flow cytometry shows that OP-G2 IgG can bind to nonactivated platelets. Quantitative binding assays indicate that nonactivated platelets bind approximately 50,000 125I-OP-G2 molecules/platelet. Furthermore, the affinity of OP-G2 for platelets activated with thrombin is roughly fivefold higher (nonactivated, kd = 24.8 nmol/L; activated, kd = 4.9 nmol/L). These results suggest that the RGD recognition site of GPIIb-IIIa is available to macromolecules that contain RGD even on nonactivated platelets, provided that the affinity of the ligand is adequate.  相似文献   

11.
Fibrinogen, fibronectin, von Willebrand factor, and thrombospondin are four large glycoproteins that bind to thrombin-stimulated platelets and influence cellular adhesive functions. The effects of five monoclonal antibodies that react with platelet membrane glycoproteins (GP) IIb and/or IIIa on the binding of these four molecules to stimulated platelets were assessed. Tab and PMI-1, antibodies recognizing GPIIb, had no effect, whereas 10E5 and 2G12, antibodies that immunoprecipitate both GPIIb and IIIa in the presence of calcium, inhibited binding of all four ligands by greater than 85%. T10, an antibody specific for the GPIIb-IIIa complex, produced partial inhibition (60% to 80%) of the binding of each ligand. Inhibitory antibodies were effective in the same dose range for all four proteins and also inhibited binding of fibrinogen, fibronectin, and von Willebrand factor to receptors fixed in an induced state (thrombin-stimulated platelets fixed with paraformaldehyde). Thrombospondin did not bind to these fixed cell preparations. The results suggest that these four adhesive proteins have a related mechanism of binding to thrombin-stimulated platelets. This related mechanism may entail the sharing of some, but not necessarily all, binding sites for the four ligands or a proximal relationship between these binding sites.  相似文献   

12.
Legrand  C; Dubernard  V; Nurden  AT 《Blood》1989,73(5):1226-1234
Affinity purified anti-fibrinogen (anti-Fg) Fab fragments were used to study the mechanism of expression of alpha-granule fibrinogen on activated platelets. Low amounts of the radiolabeled anti-Fg Fab bound to unstimulated or adenosine diphosphate (ADP)-stimulated cells. They readily bound to platelets stimulated with collagen, alpha-thrombin or gamma-thrombin in the presence of divalent cations. At 1 n mol/L alpha- thrombin or 25 nmol/L gamma-thrombin, platelet fibrinogen was expressed on the surface of the cells notwithstanding the presence of AP-2, a monoclonal antibody to the glycoprotein (GP) IIb-IIIa complex, or the synthetic peptides Arg-Gly-Asp-Ser and gamma 400-411, all substances that prevented the binding of plasma fibrinogen to platelets. These results suggest that platelet fibrinogen may interact with its receptors during its translocation from the alpha-granules to the plasma membrane and, thus, not occupy the same sites as those available for plasma fibrinogen on the surface of the cell. Furthermore, we found that platelet fibrinogen was expressed on the thrombin-stimulated platelets of a Glanzmann's thrombasthenia variant that failed to bind plasma fibrinogen. Normal platelets stimulated with 5 nmol/L alpha- thrombin bound increased amounts of the anti-fg Fab, the additional expression being inhibited by the anti-GP IIb-IIIa monoclonal antibody or by Gly-Pro-Arg-Pro, an inhibitor of fibrin polymer formation. This suggests that rebinding to externally located GP IIb-IIIa complexes becomes important once fibrin is formed.  相似文献   

13.
Platelet glycoprotein (GP) IIb-IIIa inhibitors may become useful antithrombotic agents. Ro 43-5054 is a low molecular weight, noncyclic, peptidomimetic inhibitor that is three orders of magnitude more potent than RGDS in inhibiting fibrinogen binding to purified GPIIb-IIIa and in preventing platelet aggregation. Comparisons of RGDS and Ro 43-5054 in cell adhesion assays showed that, in contrast to RGDS, Ro 43-5054 was highly selective GPIIb-IIIa inhibitor. Effects of RGDV and Ro 43-5054 on the conformation and activation state of GPIIb-IIIa were also examined. RGDV and Ro 43-5054 induced conformational changes in purified inactive GPIIb-IIIa as determined by binding of the monoclonal antibody D3GP3 (D3). These conformational alterations were not reversed after inhibitor removal, as indicated by the continued exposure of the D3 epitope and a newly acquired ability to bind fibrinogen. Similarly, RGDV and Ro 43-5054 induced conformational changes in GPIIb-IIIa on the intact platelet. However, after removal of the inhibitors, exposure of the D3 epitope was fully reversed and the platelets did not aggregate in the absence of agonist. Thus, while RGD(X) peptides and Ro 43-5054 transformed purified inactive GPIIb-IIIa into an irreversibly activated conformer, the effects of these inhibitors were reversible on the intact platelet. This suggests that factors present in the platelet membrane or cytoplasm may regulate in part the ability of the complex to shift between active and inactive conformers.  相似文献   

14.
We found that a monoclonal antibody to the glycoprotein (GP) IIb-IIIa complex, PMA4, induces fibrinogen binding to platelets, and we examined the mechanism involved. Affinity chromatography and crossed immunoelectrophoresis showed that PMA4 recognized an epitope on the GPIIb-IIIa complex-specific domain. The binding of 125I-fibrinogen to platelets was induced by PMA4 in a concentration-dependent manner and was blocked by EDTA, RGDS peptides and an anti-GPIIb-IIIa monoclonal antibody, PMA1. Binding of the divalent antibody to the GPIIb-IIIa complex was necessary to induce fibrinogen binding and subsequent platelet aggregation, since Fab fragments, unlike PMA4 IgG and F(ab')2 fragments, did not induce fibrinogen binding or aggregation. The PMA4 IgG induced fibrinogen binding, serotonin secretion, and Ca2+ mobilization, whereas F(ab')2 induced fibrinogen binding only. In addition, F(ab')2-induced fibrinogen binding was not abolished in the presence of aspirin, H-7, a protein kinase C inhibitor, PGE1 or dibutyryl cyclic AMP. These results demonstrate that the binding of PMA4 divalent molecules to the GPIIb-IIIa complex can expose platelet fibrinogen receptors in the absence of the stimulatory effects of intracellular mediators on platelets. Thus, we conclude that the fibrinogen receptors on the GPIIb-IIIa complex can be exposed by direct action of the antibody on the complex molecules.  相似文献   

15.
OBJECTIVE: To evaluate the clinical significance of factor XIII (FXIII) in primary antiphospholipid syndrome (APS). METHODS: A cross-sectional study including patients with primary APS (n =29), persistent carriers of idiopathic antiphospholipid antibodies (aPL) with no history of thrombosis (n = 14), thrombotic patients with inherited thrombophilia (n =24), healthy controls (n =28), and patients with mitral and aortic valve prosthesis (n =32, as controls for FXIII only). FXIII and fibrinogen were measured by functional assays: IgG anticardiolipin antibody (aCL), IgG anti-beta2-glycoprotein I (anti-beta2-GPI), and plasminogen activator inhibitor (PAI) by immunoassay; and paraoxonase activity by paranitrophenol formation. Intima-media thickness (IMT) of carotid arteries was determined by high resolution sonography. RESULTS: FXIII activity (FXIIIa) was highest in primary APS (p= 0.001), particularly in patients with multiple occlusions (n =12) versus those with single occlusion (158 +/- 45% vs 118 +/- 38%; p=0.02). In primary APS, FXIII positively correlated with PAI (p=0.003) and fibrinogen (p = 0.005). Similarly in the thrombotic control group, FXIIIa correlated with PAI (p =0.05) and fibrinogen (0.007). In primary APS, FXIIIa was related to the IMT of all carotid artery segments (p always < 0.01). In thrombotic controls FXIIIa correlated only to the IMT of the common carotid (p =0.01). In primary APS, FXIIIa was strongly associated with IgG aCL and IgG anti-beta2-GPI (p=0.005 for both). These associations were weaker in the aPL group (FXIIIa with IgG aCL, p= 0.02, with IgG anti-beta2-GPI, p=0.04). CONCLUSION: Enhanced FXIII activity may contribute to atherothrombosis in primary APS via increased fibrin/fibrinogen cross-linking. This pathway is not exclusive to primary APS, being present also in thrombotic controls, but the presence of IgG aPL may favor a higher degree of FXIIIa activation in the primary APS group.  相似文献   

16.
Mondoro  TH; Wall  CD; White  MM; Jennings  LK 《Blood》1996,88(10):3824-3830
Ligand-induced binding sites (LIBS) are neoantigenic regions of glycoprotein (GP)IIb-IIIa that are exposed upon interaction of the receptor with the ligand fibrinogen or the ligand recognition sequence (RGDS). LIBS have been suggested to contribute to postreceptor occupancy events such as full-scale platelet aggregation, adhesion to collagen, and clot retraction. This study examined the induction requirements of a GPIIIa LIBS with regard to ligand specificity. Through the use of the anti-LIBS D3, we report that this complex- activating antibody induces fibrinogen- and von Willebrand factor- binding to GPIIb-IIIa on intact platelets. Bound ligand was detected by flow cytometric analysis and platelet aggregation assays. These bound ligands increased the number of D3-binding sites and altered the affinity of D3 for GPIIb-IIIa on platelets. In contrast, activation of platelet GPIIb-IIIa by D3 did not increase the binding of another RGD- containing ligand, vitronectin. Furthermore, bound vitronectin on thrombin-stimulated platelets did not cause the expression of the D3 LIBS epitope. We conclude direct activation of GPIIb-IIIa in the absence of platelet activation results in selective ligand interaction and that D3 LIBS induction requires the binding of the multivalent ligands, fibrinogen or von Willebrand factor. Thus, the region of GPIIIa recognized by D3 may be an important regulatory domain in ligand- receptor interactions that directly mediate platelet aggregation.  相似文献   

17.
Plasma factor XIII (FXIII) is a tetrameric zymogen consisting of two potentially active A subunits (FXIII-A) and two carrier/inhibitory B subunits (FXIII-B). In the final phase of the coagulation cascade, FXIII is converted into an active transglutaminase (FXIIIa) by thrombin and Ca (2 + ). FXIIIa strengthens fibrin clot mechanically by cross-linking fibrin chains. In addition, FXIIIa is a key regulator of fibrinolysis, protecting newly formed fibrin from the fibrinolytic machinery by binding α (2)-plasmin inhibitor to the fibrin meshwork. FXIII is essential for maintaining hemostasis; its severe deficiency causes a life-threatening bleeding diathesis. The involvement of FXIII in thrombotic diseases and its association with the risk of these disorders is less clear. The role of FXIII in atherothrombotic diseases has been recently reviewed. This article offers a general overview of the relationship between FXIII and venous thromboembolism (VTE), to collect individual publications on this topic, present conclusions, and examine limitations of published studies. Special attention is given to the association of FXIII-A polymorphism with the risk of VTE, which has provoked considerable interest over the last decade.  相似文献   

18.
Nievelstein  PF; Sixma  JJ 《Blood》1988,72(1):82-88
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.  相似文献   

19.
E I Peerschke 《Blood》1992,79(4):948-953
Platelets contain a pool of endogenous adhesive proteins that can be released and may bind to surface membrane receptors under appropriate conditions. Because the binding of exogenous fibrinogen to platelets was shown previously to be accompanied by a time-dependent decrease in fibrinogen accessibility to antibody and enzymes, studies were performed to evaluate changes in the expression of endogenous fibrinogen released from thrombin-stimulated platelets using monospecific polyclonal and monoclonal antibody F(ab')2 fragments. Parallel studies were performed to compare the expression of released fibronectin and von Willebrand factor (vWF). Binding of polyclonal antibody F(ab')2 fragments directed against individual adhesive proteins was inhibited by EDTA or the 10E5 monoclonal antibody, suggesting that fibrinogen, fibronectin, and vWF expression was mediated, in large part, by divalent cation-dependent interactions with the glycoprotein IIb-IIIa complex. Interestingly, when polyclonal antibody F(ab')2 fragments were added to platelet suspensions at discrete times after thrombin stimulation, antifibrinogen F(ab')2 binding decreased by 72% +/- 15% (mean +/- SD, n = 22) over a 60-minute time course, whereas antifibronectin and anti-vWF antibody F(ab')2 fragment binding changed minimally (6% +/- 23%, n = 22 and 3% +/- 26%, n = 14, respectively). Similar observations were made with monoclonal antibodies. Parallel experiments using 125I-labeled fibrinogen as a marker indicated that the observed decrease in antifibrinogen F(ab')2 binding was not accompanied by fibrinogen dissociation. Moreover, antibody accessibility to platelet-bound fibrinogen could be restored after Triton X-100 platelet lysis. The data suggest that fibrinogen, fibronectin, and vWF are not coordinately expressed on thrombin-stimulated platelets. Rather, fibrinogen expression appears transient compared with the expression of fibronectin and vWF. The ability of platelets to secrete and organize adhesive proteins on their surface is likely to have important implications for hemostasis and thrombosis.  相似文献   

20.
《Platelets》2013,24(4):190-196
In this study, we have examined whether the platelet Fc-receptor, FcγRII (CD32), is associated with either of the two major platelet membrane glycoproteins, the GPIb-IX complex and the GPIIb-IIIa complex. Monoclonal and polyclonal anti-GPIb-IX complex antibodies inhibited to only a moderate degree (< 40%) the binding of the anti-FcγRII monoclonal antibody, IV.3, to platelets. In contrast, 6 of 12 anti-GPIIb-IIIa monoclonal antibodies and a polyclonal, affinity-purified rabbit anti-GPIIb-IIIa antibody strongly cross-blocked the binding of IV.3 to platelets. This inhibition was dependent upon the Fab-mediated binding of these antibodies to the GPIIb-IIIa complex since they did not inhibit the binding of IV.3 to Glanzmann's thrombasthenic platelets which have normal levels of FcγRII but lack the GPIIb-IIIa complex. The anti-GPIIb-IIIa monoclonal antibodies, AP3 and VM16a, had no effect on platelet aggregation induced by ADP or thrombin but inhibited Fc-receptor-dependent platelet aggregation as induced by either acetone-aggregated human IgG or by activating monoclonal antibodies against GPIV, PTA1 or CD9. F(ab')2 fragments of these two anti-GPIIb-IIIa monoclonal antibodies also inhibited Fc-receptor-dependent platelet aggregation indicating that the observed interference by intact antibody was not due to the direct interaction of the Fc-portion of the antigen-antibody complex with FcγRII. In addition, the inhibitory anti-GPIIb-IIIa antibodies cross-blocked the binding of IV.3 to platelets at 0°C as well as at 22°C suggesting that the observed inhibition was not dependent on the lateral mobility of either GP IIb-IIIa or FcγRII in the platelet membrane. The combined results therefore strongly suggest that the platelet Fc-receptor, FcγRII, is topographically associated with the GPIIb-IIIa complex in the intact platelet membrane.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号