Monocytes Down-Regulate Platelet Activation Induced by a Collagen Surface.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1788-1788
Author(s):  
Bertrand Arnaud ◽  
Veronique Ollivier ◽  
Jamel El Benna ◽  
Martine Jandrot-Perrus ◽  
Nadine Ajzenberg

Abstract Damage of blood vessels exposes the subendothelial matrix and results in the adhesion of platelets and monocytes at the site of injury. On eroded atherosclerotic plaques, platelets avidly bind to abundant type I collagen and monocytes are recruited by activated platelets. The purpose of the current investigation was to examine reciprocal interaction between platelets and monocytes upon exposure to a collagen surface. CD14 isolated monocytes and washed platelets were incubated separately or together in a 1/100 ratio in plates coated with type I fibrillar collagen. Platelet activation was assessed by measuring P-selectin expression by flow cytometry and RANTES secretion by ELISA. Platelet adherence and activation on immobilized collagen was analysed by confocal microscopy using FITC-phalloidin. Alternatively, cell-cell contacts were prevented by incubating platelets and monocytes in transwell coculture dishes, both parts of which were coated with collagen. In selected experiments cells were pretreated with the anti-PECAM 1.3 monoclonal antibody or with L-NMMA (NG-methyl-L-arginine, inhibitor of NO synthesis). IL8 was measured as an activation marker of monocytes. In co-incubation studies, collagen-activated platelets triggered IL8 secretion (6-fold increased) by monocytes, in agreement with previous observations indicating that platelets adherent to collagen delivers activating signals to monocytes. We have then focused our attention on the effect of monocytes on platelet activation. Unexpectedly, when monocytes were added five minutes after platelets to the collagen-coated plates, we obtained a decreased platelet expression of P-selectin by 42% (15.2 ± 9.1% positive platelets versus 26.3 ± 11.7% in the absence of monocytes, p = 0.0053, n = 16) and RANTES secretion by 39% (p < 0.0001, n = 6). The inhibitory effect of monocytes on platelet secretion decreased when the time to which they were added to platelets and collagen increased. Platelets incubated with immobilized collagen adhered and formed large aggregates consistent with a strong activation state. When monocytes were added, they established contacts with platelets while the number and size of the aggregates were dramatically decreased and isolated platelets were observed. In experiments performed in transwell coculture dishes, platelet P-selectin expression and RANTES secretion returned to the levels obtained in the absence of monocytes indicating that cell-cell contacts were required to inhibit platelet secretion induced by collagen. Preincubation of monocytes with anti-PECAM 1.3 reduced the inhibition of collagen-induced P-selectin expression and of RANTES secretion by ~ 40 %. Moreover, the inhibitory effect of monocytes on platelet aggregation appeared to be reversed by the anti-PECAM 1.3 antibody with a loss of individual platelets and the presence or large aggregates. In the presence of L-NMMA pre-treated monocytes, RANTES secretion was similar to the value obtained in the absence of monocytes. Together, our data provide evidence that, monocytes limit the initial phase of platelet activation by a collagen surface. The mechanism of this effect is dependent on cell-cell contacts. It is, at least in part, mediated by PECAM-1 with a contribution of NO. The interaction of platelets with monocytes at the surface of a damaged vessel would thus have two different effects:it would limit platelet activation and recruitment andit would increase the contribution of monocytes in inflammatory and procoagulant responses.

1984 ◽  
Vol 99 (6) ◽  
pp. 2140-2145 ◽  
Author(s):  
C R Ill ◽  
E Engvall ◽  
E Ruoslahti

The binding of platelets to components in the subendothelial matrix is an initial event in hemostasis and thrombosis. The glycoprotein components of the matrix are considered important in this interaction. Of these, collagen binds and activates platelets and induces their aggregation. In this study we demonstrate that substrate-bound laminin causes time- and concentration-dependent adherence of human platelets to the substrate. The binding of platelets to laminin was found to be similar in some respects, but different in others, to their binding to surfaces coated with fibronectin or collagen. The binding of platelets to laminin or fibronectin was not associated with their activation under conditions in which type I collagen activates the platelets as measured by [14C]serotonin secretion. Platelets bound to laminin and fibronectin differed in their appearance; they remained rounded on laminin whereas they flattened completely on fibronectin. Binding of platelets to fibronectin, but not laminin, is inhibited by a recently described peptide (Pierschbacher, M., and E. Ruoslahti, 1984, Nature (Lond.), 309:30-33) containing the cell-attachment tetrapeptide sequence of fibronectin, which suggests that separate receptors exist for laminin and fibronectin. These studies establish laminin as a platelet-binding protein and suggest that laminin can contribute to the adhesiveness of exposed tissue matrices to platelets. Since laminin and fibronectin do not activate platelets, whereas collagen does, and laminin differs from fibronectin in that it does not induce spreading of the attached platelets, all three proteins appear to confer different signals to the platelets. Some of these may be related to platelet functions other than those necessary for the formation of a hemostatic plug.


Blood ◽  
1993 ◽  
Vol 82 (7) ◽  
pp. 1985-1988 ◽  
Author(s):  
F Fauvel-Lafeve ◽  
V Tabaka ◽  
JP Caen ◽  
YJ Legrand

Abstract Bernard-Soulier syndrome (BSS) platelets, which lack the membrane glycoprotein complex Ib-IX, do not adhere to subendothelium. The adhesion of platelets from two patients with BSS to subendothelial microfibrils (MFs) and type I collagen was compared in an in vitro assay adapted to patients with low platelet count. With both patients, platelet adhesion to MFs was strongly defective, whereas the adhesion to collagen was normal. The involvement of GPIb in the MFs-induced platelet adhesion was confirmed by the inhibitory effect of a MoAb (AN51) to the von Willebrand (vWF) factor binding domain of GPIb. The adhesion of platelets to MFs thus requires GPIb-IX and an axis MFs-vWF- GPIb-IX seems therefore to be prevalent in the reactivity of platelets with subendothelium.


Blood ◽  
1993 ◽  
Vol 82 (7) ◽  
pp. 1985-1988
Author(s):  
F Fauvel-Lafeve ◽  
V Tabaka ◽  
JP Caen ◽  
YJ Legrand

Bernard-Soulier syndrome (BSS) platelets, which lack the membrane glycoprotein complex Ib-IX, do not adhere to subendothelium. The adhesion of platelets from two patients with BSS to subendothelial microfibrils (MFs) and type I collagen was compared in an in vitro assay adapted to patients with low platelet count. With both patients, platelet adhesion to MFs was strongly defective, whereas the adhesion to collagen was normal. The involvement of GPIb in the MFs-induced platelet adhesion was confirmed by the inhibitory effect of a MoAb (AN51) to the von Willebrand (vWF) factor binding domain of GPIb. The adhesion of platelets to MFs thus requires GPIb-IX and an axis MFs-vWF- GPIb-IX seems therefore to be prevalent in the reactivity of platelets with subendothelium.


2020 ◽  
Author(s):  
Hitomi Fujisaki ◽  
Sugiko Futaki ◽  
Masashi Yamada ◽  
Kiyotoshi Sekiguchi ◽  
Toshihiko Hayashi ◽  
...  

AbstractIn culture system, environmental factors, such as increasing exogenous growth factors and adhesion to type I collagen (Col-I) induce epithelial-to-mesenchymal transition (EMT) in cells. Col-I molecules maintain a non-fibril form under acidic conditions, and they reassemble into fibrils under physiological conditions. Col-I fibrils often assemble to form three-dimensional gels. The gels and non-gel-form of Col-I can be utilized as culture substrates and different gel-forming state often elicit different cell behaviors. However, gel-form dependent effects on cell behaviors, including EMT induction, remain unclear. EMT induction in lung cancer cell line A549 has been reported via adhesion to Col-I but the effects of gel form dependency are unelucidated. This study investigated the changes in EMT-related behaviors in A549 cells cultured on Col-I gels.We examined cell morphology, proliferation, single-cell migration and expression of EMT-related features in A549 cells cultured on gels or non-gel form of Col-I and non-treated dish with or without transforming growth factor (TGF)-β1. On Col-I gels, some cells kept cell–cell contacts and formed clusters, others maintained single-cell form. In cell–cell contact regions, E-cadherin expression was downregulated, whereas that of N-cadherin was upregulated. Vimentin and integrins α2 and β1 expression were not increased. In TGF-β1-treated A549 cells, cadherin switched from E- to N-cadherin. Their morphology changed to a mesenchymal form and cells scattered with no cluster formation. Vimentin, integrins α2 and β1 expression were upregulated. Thus, we concluded that culture on Col-I fibrous gels induced E- to N-cadherin switching without other EMT-related phenotypes in A549 cells.


1988 ◽  
Vol 60 (03) ◽  
pp. 514-517 ◽  
Author(s):  
Perumal Thiagarajan ◽  
Kathleen Kelly

SummaryPlatelets adhere to vitronectin substrate following activation with physiological concentrations of thrombin. Adhesion of activated platelets to vitronectin substrate is dependent upon the presence of divalent cations, the amount of vitronectin, and the duration of adhesion assay. The adhesion of platelets is inhibited by synthetic peptides containing the sequence of Arg-Gly-Asp. In addition, monoclonal antibodies to glycoprotein IIb-IIIa complex inhibit the adhesion of activated platelets to vitronectin substrate in a dose-dependent manner. These studies suggest that the glycoprotein IIb-IIIa complex on activated platelets may interact with vitronectin substrate through the Arg-Gly-Asp mechanism. Since vitronectin is present in the subendothelial matrix, it might be involved in platelet-vessel wall interactions


2000 ◽  
Vol 350 (3) ◽  
pp. 777-783 ◽  
Author(s):  
Jean-Claude MONBOISSE ◽  
Laure RITTIE ◽  
Hasnae LAMFARRAJ ◽  
Roselyne GARNOTEL ◽  
Philippe GILLERY

Glycation and glycoxidation processes, which are increased in diabetes mellitus, are generally considered causative mechanisms of long-term complications. With reference to our previous studies, type-I and -IV collagens could induce differentially the adhesion and stimulation of polymorphonuclear leucocytes (PMNs). As PMNs play a role in sustained diabetic oxidative stress, the present study was designed to determine whether in vitro glycoxidation of these macromolecules could alter PMN adhesion, activation and migration. The adhesion of PMNs to in vitro-glycoxidized collagens was significantly increased when compared with control collagens: +37% (P < 0.05) and +99% (P < 0.01) for collagens I and IV, respectively. Glycoxidized type-I collagen increased the chemotactic properties of PMNs without significant stimulatory effect on respiratory burst, whereas pre-incubation of PMNs with glycoxidized type-I collagen induced a priming on subsequent stimulation by N-formyl-methionyl-leucyl-phenylalanine. Glycoxidation of type-IV collagen suppressed its inhibitory effect on further PMN stimulation or migration. Collectively, these results indicate that glycoxidation of two major extracellular-matrix collagens considerably alters their ability to modulate PMN migration and production of reactive oxygen species. This imbalance in PMN metabolism may be a major event in the increased oxidative status that characterizes diabetes mellitus.


2014 ◽  
Vol 2014 ◽  
pp. 1-7 ◽  
Author(s):  
Chen Qi ◽  
Xu Xiaofeng ◽  
Wang Xiaoguang

Objective. To investigate the effects of Toll-like receptors in stem cell osteogenesis.Methods. Bone marrow mesenchymal stem cells (BMSCs) were divided into the blank group, the TLR-3 activated group, and the TLR-4 activated group. After 10 days’ osteogenic-promoting culture, expression of type I collagen and osteocalcin was determined by Western blot. Osteoblasts (OBs) were also divided into three groups mentioned above. Alkaline phosphatase (ALP) and alizarin red staining were performed after 10 days’ ossification-inducing culture. The expression ofβ-catenin was investigated by Western blot.Results. Both the TLR-3 and TLR-4 activated groups had increased expression of type I collagen and osteocalcin; the effect of TLR-4 was stronger. The intensity of alizarin red and ALP staining was strongest in the TLR-3 activated group and weakest in the TLR-4 activated group. Activation of TLR-4 decreased the expression ofβ-catenin, whilst activation of TLR-3 did not affect the expression ofβ-catenin.Discussion. This study suggested that both TLR-3 and -4 promoted differentiation of BMSCs to OBs. TLR-3 had an inducing effect on the ossification of OBs to osteocytes, whilst the effect of TLR-4 was the opposite because of its inhibitory effect on the Wnt signaling pathway.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1050-1050
Author(s):  
Angela Doerr ◽  
Denise Pedrosa ◽  
Maria Schander ◽  
Yotis A. Senis ◽  
Alexandra Mazharian ◽  
...  

Abstract Background Thrombus formation is a complex, dynamic and multistep process, based on two crucial steps: platelet adhesion and platelet aggregation that both involve the large multimeric plasma glycoprotein Von Willebrand Factor (VWF). VWF binding to the GPIb/X/V complex initiates platelet adhesion to the vessel wall at high shear stress and triggers platelet activation resulting in the generation of thrombin and activation of integrin αIIbβ3 on the platelet surface. This activation of αIIbβ3 in turn leads to outside-in signalling and promotes binding of αIIbβ3 to fibrinogen and VWF, mediating thrombus growth. Trigging receptor expressed on myeloid cells like transcript-1 (TLT-1) is a transmembrane receptor, which is targeted to α-granules of platelets and megakaryocytes. Thrombin-induced platelet activation rapidly presents TLT-1 on the platelet surface and releases a soluble form (sTLT-1) into the circulation. To date the only known ligand for TLT-1 is fibrinogen and TLT-1 has been implicated in the regulation of inflammation-associated thrombosis. Interestingly, a putative interaction of VWF with TLT-1 was indicated by a screen with known platelet receptors. Aim We aimed to evaluate the effect of TLT-1/VWF interaction on platelet aggregation and thrombus formation. Methods Recombinant TLT-1 and VWF were purified and the interaction between TLT-1 and VWF was analyzed by surface plasmon resonance. Static interaction was confirmed by an ELISA based binding assay. Flow assays assessed TLT-1 dependent thrombus formation in vitro. The effects of TLT-1 knockout on thrombus formation in vivo were examined via intravital microscopy of the flow restricted inferior vena cava (IVC) and imaging of platelet attachment and fibrin formation over 6 hours. Furthermore, thrombus formation and resolution was followed by high resolution ultrasound imaging after stenosis induction for 28 days. Integrin aIIbb3 activation was analysed by flow cytometry using the JonA antibody in murine platelet rich plasma. Results VWF bound to soluble TLT-1 with high affinity in a calcium dependent manner (K D = 1.9 nM). The binding site on VWF was mapped to the A3D4 domains and high molecular weight VWF multimers had the greatest affinity for TLT-1. Moreover, HEK293 cells transfected with TLT-1 bound to VWF and VWF strings formed specifically on TLT-1 expressing cells, confirming the interaction between the two proteins. VWF inhibited the binding of fibrinogen to TLT-1, suggesting that VWF is a preferred binding partner of TLT-1. Human platelets exhibited increased TLT-1 surface expression after TRAP-6 induced platelet activation and TLT-1 was detected throughout thrombi formed under flow. Furthermore, a TLT-1 blocking antibody inhibited the interaction of TLT-1 with VWF and reduced platelet capture to type I collagen under shear stress. Ex vivo perfusion of blood from TLT-1 knock out mice over type I collagen also resulted in reduced thrombus formation compared to blood from wild-type mice. TLT-1 knock-out platelets were activated by thrombin similar to wild-type controls, based on P-selectin expression in platelet rich plasma. However, activation of integrin αIIbβ3 determined by JonA staining was reduced in the absence of TLT-1. This phenotype of reduced integrin αIIbβ3 activation on P-selectin positive platelets was phenocopied by the thrombin platelet response in platelet rich plasma from VWF -/- mice, but not GPIbα-deficient mice, indicating that the TLT-1-VWF interaction on platelets directly influences integrin αIIbβ3 activation. Significantly, thrombus formation was markedly reduced in TLT-1 knockout mice in the IVC model in vivo in comparison to wild-type mice. Conclusions This study demonstrates that TLT-1 is a novel platelet ligand for VWF, and that TLT-1 may preferentially bind VWF over fibrinogen. We propose a TLT-1/VWF dependent integrin αIIbβ3 activation mechanism which plays a pivotal role in thrombus formation under non-inflammatory and potentially inflammatory conditions. Disclosures Ruf: ICONIC Therapeutics: Consultancy; MeruVasimmune: Current holder of individual stocks in a privately-held company; ARCA bioscience: Consultancy, Patents & Royalties.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1437-1437
Author(s):  
Matthew F Kee ◽  
Yongzhi Qiu ◽  
David R Myers ◽  
Yumiko Sakurai ◽  
Wilbur A Lam

Abstract Background: Vascular injury causes platelets to initiate hemostasis by first adhering to exposed subendothelial matrix proteins such as collagen. While the biochemical and biological aspects of platelet adhesion via collagen and von willebrand factor are well characterized, if and whether the mechanical properties of the subendothelial matrix affect platelet function is relatively unknown. As purely mechanical cues, such as substrate stiffness, from collagen matrices are sensed and transduced by endothelial cells to alter their physiological processes, platelets may also exhibit similar behavior (Chein S., AM J Physiol Heart Circ Physiol, 2007). In addition, recent reports demonstrate that the stiffness of the subendothelium varies in different disease states, including atherosclerosis and changes associated with aging (Stroka KM & Aranda-Espinoza H, Blood, 2011). Therefore, understanding the effect of subendothelium stiffness on platelet adhesion, spreading, and activation will provide insight into how biomechanical and biochemical factors interact during clot formation in health and disease. To that end, we fabricated polyacrylamide (PA) gels of varying stiffnesses covalently conjugated with collagen to model the biomechanical properties of the subendothelium. Via differential crosslinking, the stiffness of these PA gels can be varied while maintaining constant collagen concentrations on the gel surface, enabling decoupling of mechanical and biochemical cues (Lam, et al, Mol Cancer, 2010; Pathak A & Kumar S, P Natl Acad Sci, 2012). With these systems, we investigated how the stiffness of the underlying collagen matrix affects platelet adhesion, spreading and activation under static and flow conditions and used pharmacological cytoskeletal inhibitors to investigate the underlying mechanotransductive mechanisms. Results and Discussions: Type I collagen was covalently conjugated to the surface of PA gels with varying substrate stiffnesses (0.25 to 100 kilopascals (kPa)); the range of vessel wall stiffness in vivo. The PA gels were then incubated with 5.5 x 106 platelets/ml for 1 hour and stained with a fluorescent membrane dye. While collagen substrate stiffness did not affect platelet adhesion, determined by the number of platelets on each gel, significant differences were observed in platelet spreading area on collagen-conjugated PA gels with stiffnesses >2.5 kPa compared to PA gels of <2.5 kPa (Fig. 1A and 1B). Using PAC-1-FITC and Annexin-V-AF488 to measure platelet integrin αIIbβ3 activation and platelet phosphatidylserine (PS) exposure, respectively, we found that varying the substrate stiffness of collagen matrices did not affect αIIbβ3 activation on adherent platelets but did result in differential levels of PS exposure on adherent platelets, with increased PS exposure on stiffer PA gels (Fig. 1C). In addition, using the myosin light chain kinase (MLCK) inhibitor ML-7 and the Rho kinase inhibitor Y-27632, we observed that platelet exposure to ML-7 eliminated the substrate stiffness-mediated effect on platelet spreading as platelet spreading on PA gels stiffer than 5 kPa was decreased to the levels of that on soft PA gels of 0.5 kPa. Y-27632 exposure did not cause a similar effect, as platelet spreading was increased for all stiffness conditions (Fig. 1D). Finally, under flow conditions using a shear rate of 150 s-1, platelet adhesion in addition to platelet spreading was mediated by substrate stiffness (Fig. 1E); possibly due to weaker collagen attachment on softer substrates that cannot sufficiently resist drag forces. Conclusion: Our results indicate that platelets adhered on collagen mechanosense the stiffness of the underlying subendothelial substrate and transduce these cues into differential levels of adhesion, spreading and activation. While MLCK mediates aspects of this process, further mechanistic studies are currently being conducted. In addition, as shear stress might interact with the observed substrate stiffness-mediated phenomenon, additional experiments under flow at different shear rates are also ongoing. Disclosures No relevant conflicts of interest to declare.


1988 ◽  
Vol 90 (2) ◽  
pp. 325-333
Author(s):  
D.J. Donaldson ◽  
J.T. Mahan ◽  
G.N. Smith

Effects of the synthetic peptides, Arg-Gly-Asp-Ser (RGDS), the amino acid sequence representing the fibroblast attachment site in fibronectin, and Arg-Gly-Glu-Ser (RGES), on collagen- and fibronectin-mediated migration in newt epidermal cells were compared. When RGDS at 50 micrograms ml-1 was included in the incubation medium of skin explants, migration in fibronectin-coated dishes was almost totally blocked. In type I collagen-coated dishes, this concentration of RGDS also inhibited migration, but to a lesser degree than on fibronectin. With 250 micrograms ml-1 of RGES in the medium, the reverse was true. Here, migration on collagen was practically non-existent, while migration on fibronectin was affected only moderately. Collagen-mediated migration was sensitive to RGDS even when the peptide was added after migration on the coated substratum was well underway. At a coating concentration of 10 micrograms ml-1 CB3, a cyanogen bromide fragment of the collagen alpha 1(I) chain, which contains no RGD sequences, was as good a migration substratum as intact collagen applied at the same coating concentration. At lower concentrations intact collagen was somewhat better than equivalent concentrations of CB3. The presence of RGDS in the medium throughout an experiment inhibited migration in CB3-coated dishes in a manner similar to its effect in dishes coated with collagen. On both substrata there appeared to be a peptide-sensitive and a peptide-insensitive component to migration. The inhibitory effect of RGES on CB3-mediated migration was also similar to its effect in collagen-coated dishes.(ABSTRACT TRUNCATED AT 250 WORDS)


Sign in / Sign up

Export Citation Format

Share Document