scholarly journals Defective adhesion of blood platelets to vascular microfibrils in the Bernard-Soulier syndrome

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.

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.


1968 ◽  
Vol 20 (03/04) ◽  
pp. 384-396 ◽  
Author(s):  
G Zbinden ◽  
S Tomlin

SummaryAn in vitro system is described in which adhesion of blood platelets to washed and tannic acid-treated red cells was assayed quantitatively by microscopic observation. ADP, epinephrine and TAME produced a reversible increase in platelet adhesiveness which was antagonized by AMP. With Evans blue, polyanetholsulfonate, phthalanilide NSC 38280, thrombin and heparin at concentrations above 1-4 u/ml the increase was irreversible. The ADP-induced increase in adhesiveness was inhibited by sodium citrate, EDTA, AMP, ATP and N-ethylmaleimide. EDTA, AMP and the SH-blocker N-ethylmaleimide also reduced spontaneous platelet adhesion to red cells. No significant effects were observed with adenosine, phenprocoumon, 5-HT, phthalanilide NSC 57155, various estrogens, progestogens and fatty acids, acetylsalicylic acid and similarly acting agents, hydroxylamine, glucose and KCN. The method may be useful for the screening of thrombogenic and antithrombotic properties of drugs.


1987 ◽  
Author(s):  
K Kariya ◽  
Y Sawada ◽  
K Ueno ◽  
I Kudo ◽  
M Aihara ◽  
...  

We have reported that the adhesion of human formalin-fixed washed platelets (FWP) to collagen was enhanced by von Willebrand factor but inhibited by plasma fibronectin (Thromb. Res. 44, 1986). Recently, platelet factor XIII is reported to be a receptor for collagen (Saito, JBC, 261, 1986). To investigate the role of factor XIII in the interaction between platelets and collagen, effect of purified XIII or rabbit anti-XIIIa on the adhesion of FWP to collagen or on the in vitro bleeding time was studies. FWP adhesion was measured by either collagen-coated glass beads column or aggregometric method using bovine type I collagen (Ethicon Inc., Dr. Kronenthal). In vitro bleeding time was measured with Thrombostat-4000 (VDG-VONDERGOLTZ), in which citrated whole blood as a sample and ADP, CaCl2 and rat type I collagen as the reagents were used. Platelet adhesion to the collagen immobilized column (1,300 ug collagen, flow rate 5 ml/min) was not changed by the addition of purified XIII (Fibrogammin, Hoechst); the adhesion were 42.7 ± 1.7% in the presence of 1% human serum albumin, 42,0 ± 0.3%, 43,0 ± 1.4% in the presence of 1 or 2 u/ml factor XIII. Furthermore, the adhesion of FWP which was added by 1:100 rabbit anti-XIIIa was 42.3 ± 1.4% and not different from that of control rabbit serum (46.1 ± 1.3%). Similar results were also obtained with different technique using aggregometer. No significant change on in vitro bleeding time was observed after the addition of 1:100 rabbit anti-XIIIa to citrated normal blood. When the binding of factor XIII to the collagen was investigated by batch method, 17%, 23% and 54% of factor XHIa in normal plasma bound to 250, 500 and 1000 ug/ml collagen, respectively. These data suggest that factor XIII is not involved in the platelet adhesion to the type I fibrillar collagen, while factor XHIa in normal plasma binds to the collaeen.


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.


Blood ◽  
1993 ◽  
Vol 82 (2) ◽  
pp. 491-496 ◽  
Author(s):  
M Diaz-Ricart ◽  
NN Tandon ◽  
M Carretero ◽  
A Ordinas ◽  
E Bastida ◽  
...  

Abstract A parallel-plate perfusion chamber has been used to evaluate the contribution of the adhesive membrane glycoprotein CD36 (GPIV) to platelet adhesion on type I collagen in flowing whole blood at a shear rate of 800 s-1. In one series of experiments, reconstituted normal blood (hematocrit 0.4; platelet count 1.5 x 10(5)/microL) was prepared from washed red blood cells, plasma, and washed platelets that had been incubated with Fab fragments of a monospecific polyclonal anti-CD36 antibody (50 micrograms/mL, 30 minutes, 37 degrees C). Percent surface coverage of collagen-coated coverslips using reconstituted blood with antibody-blocked platelets, as compared with paired reconstituted controls (100%), was 50% at 2 minutes, 87% at 5 minutes, and 90% at 10 minutes. Further studies were performed by perfusion of whole blood from a healthy donor of the Naka-negative phenotype, whose platelets constitutively lack CD36, over collagen-coated coverslips. In this case, percent surface coverage was 55% of normal controls at 2 minutes, 76% of controls at 5 minutes, and 72% of controls at 10 minutes. In both preparations, platelets lacking functional CD36 had a statistically significant decrease (P < .005) in adhesion after 2 minutes and 10 minutes perfusion but not at 5 minutes. These results show that functional CD36 facilitates the rapid adhesion of platelets to collagen and that this effect is seen at the earliest time points of their interaction.


Life ◽  
2021 ◽  
Vol 11 (10) ◽  
pp. 1107
Author(s):  
Ana Amaral ◽  
Carina Fernandes ◽  
Anna Szóstek-Mioduchowska ◽  
Karolina Lukasik ◽  
Maria Rosa Rebordão ◽  
...  

Cathepsin G (CAT) is a protease released by neutrophils when forming neutrophil extracellular traps that was already associated with inducing type I collagen (COL1) in equine endometrium in vitro. Endometrosis is a fibrotic condition mainly characterized by COL1 deposition in the equine endometrium. The objective was to evaluate if noscapine (an alkaloid for cough treatment with anti-neoplastic and anti-fibrotic properties) would reduce COL1A2 transcription (evaluated by qPCR) and COL1 protein relative abundance (evaluated by western blot) induced by CAT in equine endometrial explants from follicular and mid-luteal phases treated for 24 or 48 h. The explants treated with CAT increased COL1 expression. Noscapine decreased COL1A2 transcription at both estrous cycle phases, but COL1 relative protein only at the follicular phase, both induced by CAT. Additionally, the noscapine anti-fibrotic action was found to be more effective in the follicular phase. The CAT treatment caused more fibrosis at the longest period of treatment, while noscapine acted better at the shortest time of treatment. Our results showed that noscapine could act as an anti-fibrotic drug in equine endometrosis by inhibiting CAT in vitro. Noscapine offers a new promising therapeutic tool for treating fibrosis as a single non-selective agent to be considered in the future.


1977 ◽  
Author(s):  
F. Fauvel ◽  
Y. J. Legrand ◽  
J. P. Caen

The interaction of blood platelets with collagen is the trigger of haemostasis and, at least partly, of thrombosis. Two types of fibrillar collagens (type I and type III), present in the vessel wall, can induce the adhesion of blood platelets. This study was designed to determine which part of their molecule is involved in that phenomenon. We then devised a quantitative test based upon differential filtration of non adhesive and adhesive 14c serotonin labeled platelets through a sepharose column for the evaluation of platelet adhesion to collagen or collagen derivatives. No differences were observed in the adhesive properties of calf-skin type I and type III collagens when the degree of multimerisation was identical. Isolated alpha 1 (I) and alpha 2 chains were not active ; but platelets adhered to reassociated (alpha .1 (I))3 trimers, not to (alpha 2)3 trimers ; in type I collagen, alpha 1 chains seems to have the most essential role for platelet adhesion. Various cyanogen bromide peptides were tested : only the 217 C-terminal amino acids alpha 1 CB6 from type I and the 149 central amino acids alpha 1 CB4 from type III were active.The adhesive properties of collagen seems then associated with chemical groupings localized in these peptides : a random folding of the entire isolated chains could make these “sites” unavailable to the platelets, whereas in the triple helix characteristic of fibrillar trimers, they would be more accessible. This could explain why fibrils and small peptides are active, while isolated chains are not.


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.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1609-1609 ◽  
Author(s):  
Isabella Pallotta ◽  
Michael L. Lovett ◽  
David L. Kaplan ◽  
Alessandra Balduini

Abstract Abstract 1609 Background. The mechanisms that regulate megakaryocytic (Mk) development within the bone marrow environment remain poorly understood. The underlying relationships between Mk maturation and bone marrow components are key factors in this process. Mk development occurs in a complex microenvironment where extracellular matrices are fundamental regulatory components. The first events occur in the osteoblastic niche and include commitment of the hemopoietic progenitor cell to Mk, arrest of proliferation and initiation of endomitosis. The second step is Mk maturation and is associated with rapid cytoplasm expansion and intense synthesis of proteins. Finally Mks, which migrate to the vascular niche, convert the bulk of their cytoplasm into multiple long processes called proplatelets that protrude through the vascular endothelium into the sinusoid lumen, where the platelets are released. Hypothesis. The hypothesis for the present work is that a complex in vitro 3D bone marrow-like environment can be used to gain fundamental mechanistic insight into cell signalling and matrix-cell interactions in the bone marrow niche related to Mk development. Methods. We propose the first 3D model for Mk function in the bone marrow environment, by refining a recently proposed bioreactor platform (Lovett et al., 2007). These bioreactors consist of 3 wells (10 mm × 15 mm × 5 mm) within a PDMS block (25 mm × 60 mm × 5 mm) which is plasma bonded to cover glass for imaging. Each bioreactor well was perfused by 23 G stainless steel needles, spanned by porous silk microtubes as blood vessel scaffolds (640 μm inner diameter), positioned approximately 500–750 μm from the bottom of the bioreactor and connected to tubing for media perfusion using a programmable syringe pump. These microtubes were prepared by dipping several times straight lengths of stainless steel wire into 10–14% (w/v) aqueous silk fibroin to obtain blood vessel scaffolds with a wall thickness of around 50 mm. Defined pore sizes of 6–8 μm were obtained by adding 6 w/t % poly(ethylene oxide) (PEO) to the silk fibroin. The perfused silk tubes comprised the vascular niche and were embedded within a cell-seeded hydrogel which comprises the osteoblastic niche. The silk microtubes were coated with a combination of fibrinogen, von Willebrand Factor, type IV collagen and SDF-1 alpha, to better establish the composition of the vascular niche. Control experiments were performed by coating silk microtubes with type I collagen. After staining human umbilical cord blood derived Mks, the cell suspension was added to the hydrogel and Mk migration was analyzed in a time-dependent manner using confocal microscopy analysis. Further, flow effluent through the vascular tubes in the bioreactor was collected at regular time intervals and platelet numbers and function were analyzed by flow cytometry and microscopy. Culture released platelets were counted as CD61+ events with the same scatter properties of human blood platelets. Results. Our results showed that Mks migrated towards the vascular microtube coated with Fibrinogen, von Willebrand Factor, type IV collagen and SDF-1. Mks were also able to complete their maturation in the proximity of the microtube by extending proplatelets. Interestingly, confocal microscopy analysis revealed that Mks were able to extend proplatelets through the vascular microtube wall and release CD61+ platelet-like particles inside the vascular microtube. Cytofluorimentric analysis demonstrated that the particles collected in the flow effluent of the vascular microtube were CD61+ cells with the same scatter properties of human peripheral blood platelets. Finally, upon coating with only type I collagen Mks did not migrate towards the vascular microtube or extend proplatelets to release platelets. Thus, by mimicking the relationship between Mks and the bone marrow environment, a model to reproduce the different steps of Mk development, such as Mk migration, proplatelet formation and platelet release, is established. This is a first significant step towards relevant systems for the study of these cellular processes in detail as well as toward potentially useful in vitro platelet production systems. Conclusions. In this work we developed a new 3D bone marrow system in vitro that could represent a new tool to understand the mechanistic basis for Mk development and function, and the diseases related to these cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 450-457 ◽  
Author(s):  
DG Moon ◽  
JE Kaplan ◽  
JE Mazurkewicz

Plasma fibronectin (Fn) has been proposed to have an antithrombotic effect, protecting against platelet and fibrinogen consumption after injury. The current study was designed to determine the effect of plasma fibronectin on collagen-induced platelet aggregation. In vitro aggregometry using an isolated homologous rat system, demonstrated a significant (P less than .05) inhibitory effect of 120 micrograms/mL Fn on platelet aggregation as induced by 60 micrograms/mL fibrillar collagen (type I). The inhibition was evidenced by a threefold increase in lag time and a significant decrease in the rate and extent of aggregation. The hypothesis was also tested using an in vivo model of collagen-induced platelet aggregation. The model used was intravenous injection of 2 mg/kg of homologous type I collagen into anesthetized Sprague-Dawley rats. Injection of collagen preincubated with 4 mg/kg Fn resulted in significantly less thrombocytopenia and fibrinogen consumption as compared with injection of collagen alone. The results of both the in vitro and in vivo studies are consistent with the proposed antithrombotic effect of plasma fibronectin.


Sign in / Sign up

Export Citation Format

Share Document