Reduced Platelet Adhesion for Blended Electrospun Meshes with Low Amounts of Collagen Type I

2021 ◽  
pp. 2100267
Author(s):  
Mahyar Sameti ◽  
Kai Clarke ◽  
Prerona Dewan ◽  
Kenyatta S. Washington ◽  
Somayeh Talebzadeh ◽  
...  
1987 ◽  
Author(s):  
Philip G de Groot ◽  
Jan A van Mourik ◽  
Jan J Sixma

We have studies the binding of von Willebrand factor (vWF) to extracellular matrices of endothelial cells and smooth muscle cells and to the vessel wall of human umbilical arteries in relation to its function in supporting platelet adhesion at high shear rates. CLB-RAg 38, a monoclonal antibody directed against vWF inhibits the binding of 125I-vWF extracellular matrices completely. The binding of 125I-vWF to subendothelium is not inhibited, because there are many different binding sites. CLB-RAg 38 inhibits platelet adhesion to extracellular matrices and subendothelium, in sofar as it is dependent on plasma vWF. CLB-RAg 38 has no effect on adhesion depending on vWF already bound to the matrix or subendothelium. CLB-RAg 38 does not inhibit binding of vWF to collagen type I and type III. Another monoclonal antibody against vWF, CLB-RAg 201, completely inhibits binding of vWF to collagen type I and type III. CLB-RAg 201 does not inhibit binding of 125I-vWF ot the extracellular matrices. CLB-RAg 201 partly inhibits platelet adhesion but this inhibition is also present when the adhesion depends on vWF already present in matrix or subendothelium, indicating that CLB-RAg 201 also inhibits the adhesion of platelets directly, this in contrast to CLB-RAg 38. The epitopes for CLB-RAg 201 and 38 were found on different tryptic fragments of vWF. These data indicate that vWF binds to subendothelium and to matrices of cultured cells by mechanism that is different from binding to collagen.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3677-3677
Author(s):  
Sungwook Choi ◽  
Seth E. Snyder ◽  
David T. Moore ◽  
Gaston Vilaire ◽  
Joel S. Bennett ◽  
...  

Abstract Platelets tether to collagen in the subendothelial matrix that is exposed by vascular damage. Collagen is a particularly important matrix component in this context, not only because it is a substrate for platelet adhesion, but because it is an agonist for platelet aggregation and secretion as well. There are two platelet collagen receptors, the immunoglobulin gene superfamily member GPVI and the integrin α2Β1. Both are involved in adhesion to exposed collagen and generate downstream activating signals. α2Β1 is widely expressed and has been implicated in hemostasis and thrombosis, as well as cancer metastasis, wound healing, and angiogenesis. In mice, α2Β1 deficiency results in decreased ex vivo platelet aggregation, but normal bleeding times. In mouse tumor models, α2Β1 blockade reduces both metastasis and angiogenesis. Humans lacking α2Β1 have a mild bleeding diathesis. Given this background, α2Β1 appears to be an appropriate target for the development of small-molecule inhibitors to serve as relatively safe anti-platelet and anti-tumor agents, either acting alone or in synergy with other anti-platelet or anti-tumor agents. We have developed two classes of small-molecule α2Β1 inhibitors. The first class is targeted against the collagen binding site located on the α2 I-domain and was designed using molecular modeling to superimpose a dipeptide scaffold onto the published crystal structure of the I-domain bound to a collagen-mimetic peptide (GFOGER). These molecules block recombinant human I-domain binding to immobilized collagen type I with IC50s as low as 10 μM. Although the molecules inhibit platelet adhesion to collagen only at higher concentrations, they readily inhibit melanoma cell adhesion to collagen mimetics. It is also noteworthy that the molecules induce platelet protein phosphorylation and potentiate platelet aggregation induced by other platelet agonists, both of which can be prevented by pre-incubating platelets with monoclonal antibodies directed against the α2 I-domain, but not against GPVI. The second class of molecules was derived from proline-substituted 2,3-diaminopropionic acids and is directed against the Β1 I-like domain, an allosteric site that regulates ligand binding. These molecules are potent inhibitors of platelet adhesion to immobilized soluble collagen type I with IC50s of 10–50 nM and inhibit the adhesion of melanoma cells to collagen mimetics with IC50s of 250–350 nM. These molecules do not inhibit platelet aggregation, nor do they inhibit I-domain binding to immobilized collagen type I, behavior consistent with binding to the Β1 I-like domain. In a murine model of ferric-chloride induced carotid thrombosis, the molecules synergize with aspirin to prevent arterial thrombosis. In summary, we have developed two classes of small molecule inhibitors that impair the interaction of collagen with the integrin α2Β1. Although both classes of inhibitors bind to α2Β1, their effects on its function are substantially different, indicating that there are multiple potential strategies for inhibiting integrin function pharmacologically. Further development of these inhibitors may lead to agents that will be clinically useful in the treatment of thrombosis and cancer.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3266-3266
Author(s):  
Yasuaki Shida ◽  
Christine Brown ◽  
Jeff Mewburn ◽  
Kate Sponagle ◽  
Ozge Danisment ◽  
...  

Abstract Abstract 3266 Von Willebrand Factor (VWF) is a large multimeric glycoprotein that mediates platelet adhesion to the damaged blood vessel wall and subsequent platelet aggregation at the site of injury. Rare mutations in the VWF A3 domain, that disrupt collagen binding, have been found in patients with a mild bleeding phenotype. However, the analysis of these aberrant VWF-collagen interactions has been relatively limited. Thus, in this study, we have developed mouse models of collagen binding mutants and analyzed the function of the A3 and A1 domains using comprehensive in vitro and in vivo approaches. All of the collagen binding variant AAs are conserved in mice. 6 loss-of-function (S1731T, W1745C, S1783A, H1786D, A1 deletion, A3 deletion) and 1 gain-of-function (L1757A) variant was generated in the context of the mouse VWF cDNA. The 4 loss-of-function missense mutants have all been described in patients with mild bleeding phenotypes. The recombinant mouse VWFs (rmVWF) were synthesized in HEK293T cells and analyzed for type I and III collagen binding in both a static assay (CBA) and a flow-based assay at 2,500s−1 in which VWF is bound to collagen on a surface, and labeled platelet adhesion is quantified. The multimer profile of all the rmVWFs was normal. The expression level of the rmVWF derived from HEK293T cells was quantified. W1745C and the A3 deletion showed significantly lower levels of expression and the A1 deletion mutant showed strong intracellular retention. In the static collagen binding assay, S1731T showed almost normal binding to collagen type I and a 50% reduction in binding to collagen type III. The other 3 missense variants, W1745C, S1783A and H1786D, showed reduced binding to both collagens I and III, and the A3 deletion mutant showed absent binding. In the in vitro flow assay, the sensitivity to detect defects in collagen binding was superior to the static assay, although the patterns of binding defects were similar. W1745C showed similar low levels of platelet adhesion to both types of collagen, while S1783A and H1786D showed a lack of platelet binding on the collagen III surface similar to the A3 deletion mutant, and a reduced binding to collagen type I similar to W1745C. The gain-of-function mutant showed consistent enhanced collagen binding and platelet adhesion in the static and flow assays, respectively. In vivo studies delivered the mVWF cDNAs with a strong liver specific promoter by hydrodynamic injection. At 7 days post-delivery, the VWF:Ag levels in the WT and collagen binding variant mice were similar, apart from the W1745C mutant, that showed 14.6% levels compared to WT. Platelet counts and multimer patterns were normal with the collagen binding variants. In vivo intravital microscopy studies were performed using the cremaster arteriolar model when VWF levels were in a physiological range. Thrombosis was induced by 10%FeCl3 applied for 3 mins. Platelets were labeled in vivo by Rhodamine 6G and the thrombus development was analyzed by spinning disc confocal microscopy. Loss-of-function mutants showed transient platelet adhesion at the site of injury, however the adhesion was unstable and vessel occlusion was not observed. Using three complementary experimental systems we have been able to confirm the collagen binding defects in this group of variant VWFs. There is a differential sensitivity to the two forms of collagen and of the three experimental systems. The A3 deletion mutant consistently resulted in the most severe phenotype while the missense mutants showed variable degrees of functional deficit. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 35 (03) ◽  
pp. 211-224 ◽  
Author(s):  
G. L. Mendolicchio ◽  
Z. M. Ruggeri

SummaryThe initiation of thrombus formation at sites of vascular injury to secure haemostasis after tissue trauma requires the interaction of surface-exposed von Willebrand factor (VWF) with its primary platelet receptor, the glycoprotein (GP) Ib-IX-V complex. As an insoluble component of the extracellular matrix (ECM) of endothelial cells, VWF can directly initiate platelet adhesion. Circulating plasma VWF en-hances matrix VWF activity by binding to structures that become exposed to flowing blood, notably collagen type I and III in deeper layers of the vessel along with microfibrillar collagen type VI in the sub endothelium. Moreover, plasma VWF is required to support platelet-to-platelet adhesion – i. e. aggregation – which promotes thrombus growth and consolidation. For these reasons, understanding how plasma VWF interaction with platelet receptors is regulated, particularly any distinctive features of GPIb binding to soluble as opposed to immobilized VWF, is of paramount importance in vascular biology.This brief review will highlight knowledge acquired and key problems that remain to be solved to elucidate fully the role of VWF in normal haemostasis and pathological thrombosis.


1989 ◽  
Vol 258 (1) ◽  
pp. 157-163 ◽  
Author(s):  
L F Morton ◽  
A R Peachey ◽  
M J Barnes

The adhesion of human and rabbit platelets to collagens and collagen-derived fragments immobilized on plastic was investigated. Adhesion appeared to be independent of collagen conformation, since similar attachment occurred to collagen (type I) in monomeric form, as fibres or in denatured state. The adhesion of human platelets was stimulated to a variable degree by Mg2+, but rabbit platelet adhesion showed little if any dependence on this cation. Collagens type I, III, V and VI were all able to support adhesion, although that to collagen type V (native) was lower than that to the other collagens. Adhesion to a series of peptides derived from collagens I and III was measured. Attachment did not require the presence of peptides in triple-helical configuration. The extent of adhesion ranged from relatively high, as good as to the intact parent collagen molecule, to little if any adhesive activity beyond the non-specific (background) level. The existence of very different degrees of activity suggests that platelet adhesion is associated with specific structural sites in the collagen molecule. Adhesion in many instances was essentially in accord with the known platelet-aggregatory activity of individual peptides. However, two peptides, alpha 1(I)CB3 and alpha 1(III)CB1,8,10,2, exhibited good adhesive activity although possessing little if any aggregatory activity. Of particular interest, despite its near-total lack of aggregatory activity, adhesion to peptide alpha 1(I)CB3 was as good as that to the structurally homologous peptide alpha 1(III)CB4, in which is located a highly reactive aggregatory site. This implies that platelet adhesion to collagen may involve sites in the collagen molecule distinct from those more directly associated with aggregation.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3638-3638
Author(s):  
Luigi De Marco ◽  
Maria Rita Cozzi ◽  
Mario Mazzucato ◽  
Monica Battiston ◽  
Martine Jandrot Perrus ◽  
...  

Abstract We have investigated signaling mediated by GPIb-IX-V, GPVI and α2β1 during platelet adhesion to collagen type I either in the presence or absence of von Willebrand factor (VWF) A1A2A3 domain under flow conditions (wall shear rate 600 or 3000 s−1). We used platelets labeled with FLUO3-AM and real-time videomicroscopy with high-speed image acquisition to analyze concurrently adhesion, translocation and calcium transients in single platelets interacting with the surface. All experiments were performed in the presence of integrillin (100 μg/ml) to block the possible involvement of αIIbβ3. Platelet adhesion and activation at 600 s−1 were similar in the absence or presence of VWF A1A2A3 domain. Calcium signaling consisted of both short lasting peaks (release from intracellular stores) and long sustained waves (transmembrane flux). In our experimental condition blockade of α2β1 or GPVI with monoclonal antibodies reduced platelet adhesion by 50% and the proportion of adherent platelets that became activated by 20–30%. Concomitant inhibition of both receptors decreased platelet adhesion by >90%. Thus, both α2β1 and GPVI participate in the initial stage of platelet adhesion to collagen type I. At the higher shear rate of 3000 s−1, platelet adhesion and activation was 40–50% less than at 600 s−1 in the absence of VWF A1A2A3 domain; addition of the latter enhanced adhesion and activation by 12 to 14-fold, and >80% of the platelets exhibited stable adhesion during the 30 s observation period. Intracellular Ca++ peaks in adherent platelets reached 2–3 μM. Blockade of GP Ib-IX-V inhibited both platelet adhesion and activation by at least 90%. Blockade of α2β1 markedly reduced the number of adhering platelets, which were mostly (>90%) translocating on the surface and exhibited only short lasting Ca++ transients. Blockade of GPVI greatly decreased the number of adherent platelets, most of which, however, were firmly attached to the surface (<15% exhibiting translocation). Calcium transients in the activated platelet consisted of short lasting peaks and rare long lasting waves reaching concentrations <1.5–2 μM. These results suggest that, on a surface of collagen type I, α2β1 is required for normal platelet arrest following tethering through the VWF A1 domain-GPIb-IX-V interaction at higher shear rates, and Ca++ signaling results from the concerted action of the two receptors reinforced by GPVI. The adhesion potential of platelets exposed to collagen, therefore, appears to depend on signaling triggered by different ligand-receptor interactions varying as a function of flow conditions.


Sign in / Sign up

Export Citation Format

Share Document