scholarly journals Novel Stenotic Microchannels to Study Thrombus Formation in Shear Gradients: Influence of Shear Forces and Human Platelet-Related Factors

2019 ◽  
Vol 20 (12) ◽  
pp. 2967 ◽  
Author(s):  
Mathew Lui ◽  
Elizabeth E. Gardiner ◽  
Jane F. Arthur ◽  
Isaac Pinar ◽  
Woei Ming Lee ◽  
...  

Thrombus formation in hemostasis or thrombotic disease is initiated by the rapid adhesion, activation, and aggregation of circulating platelets in flowing blood. At arterial or pathological shear rates, for example due to vascular stenosis or circulatory support devices, platelets may be exposed to highly pulsatile blood flow, while even under constant flow platelets are exposed to pulsation due to thrombus growth or changes in vessel geometry. The aim of this study is to investigate platelet thrombus formation dynamics within flow conditions consisting of either constant or variable shear. Human platelets in anticoagulated whole blood were exposed ex vivo to collagen type I-coated microchannels subjected to constant shear in straight channels or variable shear gradients using different stenosis geometries (50%, 70%, and 90% by area). Base wall shears between 1800 and 6600 s−1, and peak wall shears of 3700 to 29,000 s−1 within stenoses were investigated, representing arterial-pathological shear conditions. Computational flow-field simulations and stenosis platelet thrombi total volume, average volume, and surface coverage were analysed. Interestingly, shear gradients dramatically changed platelet thrombi formation compared to constant base shear alone. Such shear gradients extended the range of shear at which thrombi were formed, that is, platelets became hyperthrombotic within shear gradients. Furthermore, individual healthy donors displayed quantifiable differences in extent/formation of thrombi within shear gradients, with implications for future development and testing of antiplatelet agents. In conclusion, here, we demonstrate a specific contribution of blood flow shear gradients to thrombus formation, and provide a novel platform for platelet functional testing under shear conditions.

2021 ◽  
Vol 22 (8) ◽  
pp. 4066
Author(s):  
Patrizia Marchese ◽  
Maria Lombardi ◽  
Maria Elena Mantione ◽  
Domenico Baccellieri ◽  
David Ferrara ◽  
...  

Atherothrombosis exposes vascular components to blood. Currently, new antithrombotic therapies are emerging. Herein we investigated thrombogenesis of human arteries with/without atherosclerosis, and the interaction of coagulation and vascular components, we and explored the anti-thrombogenic efficacy of blockade of the P2X purinoceptor 7 (P2X7). A confocal blood flow videomicroscopy system was performed on cryosections of internal mammary artery (IMA) or carotid plaque (CPL) determining/localizing platelets and fibrin. Blood from healthy donors elicited thrombi over arterial layers. Confocal microscopy associated thrombus with tissue presence of collagen type I, laminin, fibrin(ogen) and tissue factor (TF). The addition of antibodies blocking TF (aTF) or factor XI (aFXI) to blood significantly reduced fibrin deposition, variable platelet aggregation and aTF + aFXI almost abolished thrombus formation, showing synergy between coagulation pathways. A scarce effect of aTF over sub-endothelial regions, more abundant in tissue TF and bundles of laminin and collagen type I than deep intima, may suggest tissue thrombogenicity as molecular structure-related. Consistently with TF-related vascular function and expression of P2X7, the sections from CPL but not IMA tissue cultures pre-treated with the P2X7 antagonist A740003 demonstrated poor thrombogenesis in flow experiments. These data hint to local targeting studies on P2X7 modulation for atherothrombosis prevention/therapy.


2020 ◽  
Author(s):  
Daria S. Morozova ◽  
Alexey A. Martyanov ◽  
Sergei I. Obydennyi ◽  
Julia-Jessica D. Korobkin ◽  
Alexey V. Sokolov ◽  
...  

AbstractInfiltration of growing thrombi by leukocytes, being the key part of the thromboinflammation, is well established in vivo. The study was aimed at the development of an ex vivo simulation of this phenomenon. Thrombus formation in anticoagulated whole blood from healthy volunteers and patients was visualized by fluorescent microscopy in parallel-plate flow chambers with fibrillar collagen type I coverslips.Moving CD66b-positive cells (granulocytes) were observed in hirudinated or recalcified blood under low wall shear rate conditions (<200 s−1). These cells crawled around thrombi in a step-wise manner with an average rate of 70 nm/s. Pre-incubation of blood with leukocyte priming agents lead to a significant increase in average cell velocity. On the contrary, leukocytes from Wiskott-Aldrich syndrome patients demonstrated a 1.5-fold lower average velocity, in line with their impaired actin polymerization.Thereby, the observed features of granulocytes crawling are consistent with the neutrophil chemotaxis phenomenon. We conclude that the proposed ex vivo experimental setting allows us to observe granulocytes activity in near-physiological conditions.


2021 ◽  
Vol 10 (22) ◽  
pp. 5349
Author(s):  
Lydie Crescence ◽  
Markus Kramberg ◽  
Martine Baumann ◽  
Markus Rey ◽  
Sebastien Roux ◽  
...  

Selatogrel, a potent and reversible antagonist of the P2Y12 receptor, inhibited FeCl3-induced thrombosis in rats. Here, we report the anti-thrombotic effect of selatogrel after subcutaneous applications in guinea pigs and mice. Selatogrel inhibited platelet function only 10 min after subcutaneous application in mice. In addition, in a modified Folts thrombosis model in guinea pigs, selatogrel prevented a decrease in blood-flow, indicative of the inhibition of ongoing thrombosis, approximately 10 min after subcutaneous injection. Selatogrel fully normalised blood flow; therefore, we speculate that it may not only prevent, but also dissolve, platelet thrombi. Thrombus dissolution was investigated using real-time intravital microscopy in mice. The infusion of selatogrel during ongoing platelet thrombus formation stopped growth and induced the dissolution of the preformed platelet thrombus. In addition, platelet-rich thrombi were given 30 min to consolidate in vivo. The infusion of selatogrel dissolved the preformed and consolidated platelet thrombi. Dissolution was limited to the disintegration of the occluding part of the platelet thrombi, leaving small mural platelet aggregates to seal the blood vessel. Therefore, our experiments uncovered a novel advantage of selatogrel: the dissolution of pre-formed thrombi without the disintegration of haemostatic seals, suggesting a bipartite benefit of the early application of selatogrel in patients with acute thrombosis.


2001 ◽  
Vol 85 (06) ◽  
pp. 1097-1103 ◽  
Author(s):  
Kjell Sakariassen ◽  
Hélène Grandjean ◽  
Claire Thalamas ◽  
Bernard Boneu ◽  
Pierre Sié ◽  
...  

SummaryA number of studies have reported conflicting data on the association of the PlA1/PlA2 polymorphism of the GPIIIa gene and coronary syndromes. We have investigated the effect of this polymorphism on experimental platelet thrombus formation in man. Forty healthy male volunteers were genotyped for the PlA1/PlA2 polymorphism. Thrombus formation was induced ex vivo by exposing a tissue factor (TF) or a collagencoated coverslip in a parallel plate perfusion chamber to native blood for 2 and 4 min. The shear rates at these surfaces were 650 and 2,600 s–1. Platelet and fibrin deposition was quantified by immunoenzymatic methods. The frequencies of PlA1/PlA1 and PlA1/PlA2 genotypes were 52.5% and 47.5%, respectively. Ex vivo deposition of fibrin on TF was not affected by the PlA1/PlA2 polymorphism. However, the ex vivo platelet deposition at 650 s–1 was higher in blood from PlA1/PlA1 individuals than in PlA1/PlA2 individuals (P = 0.008 at 4 min). On collagen, neither fibrin nor platelet deposition was significantly affected by the PlA1/PlA2 polymorphism. Platelet thrombus formation is significantly influenced by genetic variations in the GPIIIa platelet receptor. This effect depends on the blood flow properties and the nature of the thrombogenic stimulus.


1997 ◽  
Vol 20 (1) ◽  
pp. 43-50 ◽  
Author(s):  
K.W.H.J. Van Der Kamp ◽  
C.P.E. Magielse ◽  
J.M. Elstrodt ◽  
J. Van Der Meer ◽  
W. Van Oeveren ◽  
...  

The blood compatibility of ventricular assist devices developed by the Helmholtz Institute Aachen (HA-VAD's) was tested on calves. Seven calves received a non-coated HIA-VAD (control) and three a Bioline heparin coated device. The circulatory support of these HIA-VAD's lasted one week. Mechanical blood cell trauma estimated by hematocrit (Hct), hemoglobin (total Hb) and free plasma hemoglobin (free Hb) levels did not differ in either group. All HIA-VAD's in the control group remained thrombus free, except on one occasion when an inflow cannula was obstructed by a thrombus located in the tip. After circulatory support, the animals in this group seemed clinically healthy. However, thrombus formation was observed in the three heparin coated HIA-VAD's. One animal in this group died from complications after re-operation for pneumothorax on the fifth day of support, whereas the other two animals seemed clinically healthy. In these three animals, a stronge decrease in platelet numbers was measured even after 24 hours of support which recovered after 72 hours. This decrease in platelet numbers was associated with a lower degree of platelet aggregation ability stimulated by ADP (p<0.05). Fibrin(ogen) degradation products (FDP) increased significantly immediately after the implantation procedure (p<0.05). Fibrinogen levels initially decreased during the implantation procedure, but increased thereafter in both groups. The FDP levels remained high in this group, although the FDP levels in both groups were decreased after the implantation procedure. The ex vivo measured circulating heparin levels were lower in the heparin coated HIV-VAD group despite the equally administrated heparin doses in both animal groups. No differences were measured in either group with regard to white blood cell (WBC) numbers and complement hemolytic activity (CH50). Despite these hemostatic changes, no mechanical trauma could be demonstrated after seven days of circulatory support.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 114-114
Author(s):  
Alessandro Zarpellon ◽  
Patrizia Marchese ◽  
Antonella Zampolli ◽  
Grazia Loredana Mendolicchio ◽  
Zaverio M. Ruggeri

Abstract Generation of α-thrombin (FIIa) in response to vascular injury is a key mechanism influencing thrombus formation. Platelet activation by FIIa is mediated by different protease activated receptors (PARs), although the most abundant FIIa binding site on platelets, but not a substrate for proteolysis, is glycoprotein (GP) Ibα in the GPIb-IX-V complex. The functional role of GPIbα in mediating/regulation thrombin functions relative to that of different PARs remains unclear. The goal of these studies was to define how binding to GPIbα can modulate FIIa functions. In mouse platelets we replaced endogenous GPIbα with either its human wild type counterpart (huGPIbα-WT) or with huGPIbα mutated at key residues involved in thrombin binding (D277N, Y276-8-9/F). Because these two mutations resulted in an undistinguishable phenotype, they are designated collectively as huGPIbα-Mut hereon. Mice expressing huGPIbα, WT or Mut, were evaluated in intravital models of arterial thrombosis induced by a ferric chloride-induced carotid artery lesion and venous thromboembolism induced by intravenous α-thrombin injection. Moreover, the blood of huGPIbα WT or Mut mice was also tested in an ex vivo model of thrombus formation upon perfusion over a thrombogenic surface under controlled flow conditions and platelets were evaluated for their responses to FIIa-induced activation. Mice expressing huGPIbα - WT or Mut - have comparable platelet counts and GPIbα surface density. Moreover, huGPIbα-WT platelets bind FIIa similarly than their normal human control counterpart, while huGPIbα-Mut platelets have essentially no detectable FIIa binding. Upon FIIa stimulation, which on mouse platelets is mediated by PAR4, aggregation and Ca2+ transients were significantly enhanced in huGPIbα-Mut as compared to huGPIbα-WT. In contrast, blocking FIIa binding to GPIbα on human platelets essentially abolished FIIa mediated activation, which in human occurs predominantly through PAR1. These results are compatible with the conclusion that, in mice, GPIbα is a competitive inhibitor of FIIa for PAR4-mediated functions. In the presence of metabolically inactive (PGE1 treated) huGPIbα-Mut washed platelets, the clotting time of a purified fibrinogen solution was significantly shorter when triggered by relative high concentration of FIIa (4 nM), but pronouncedly prolonged at a lower FIIa concentration (0.5 nM). Clot visualization showed a much more structured fibrin mesh in the presence of huGPIbα-WT platelets, which was lost with in the presence of huGPIbα-Mut platelets. Mutant mice tested in a model of carotid artery injury exhibited a pronounced prothrombotic phenotype, with a shorter time to occlusion. However they were protected from death induced by I.V. injection of α-thrombin. In ex vivo perfusion studies, the total volume of platelet aggregates formed in huGPIbα-Mut mouse blood exposed to acid-insoluble fibrillar collagen type I was slightly bigger than in huGPIbα-WT mice, but the number of thrombi was increased and their individual size smaller. These huGPIbα-Mut platelets exhibited clear signs of increased activation, as visualized by scanning electron microscopy (SEM). Strikingly, fibrin was almost totally absent in the huGPIbα-Mut thrombi. This was in striking contrast with what observed in huGPIbα-WT mice, in which the surface of platelet thrombi with directly and tightly connected with thick fibrin fibers as visualized by scanning electron microscopy. Possibly because of the reduced platelet membrane-fibrin fibril connection in huGPIbα-Mut platelets, these mice were significantly less susceptible to death when injected with an α-thrombin dose that caused 80% mortality plus in huGPIbα-WT mice. Thus, mice whose platelets have defective α-thrombin binding to GPIbα have a prothrombotic phenotype in high shear stress flow arteries and are protected from thromboembolic death in the low shear stress venous circulation. Our findings identify GPIbα as a relevant FIIa activity modulator in hemostasis and thrombosis through distinct and opposite mechanisms affecting platelet activation (The Yin) and fibrin formation (The Yang). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 216-216
Author(s):  
Jose A. Guerrero ◽  
Taisuke Kanaji ◽  
Junling Liu ◽  
Susan Russell ◽  
T.K. Gartner ◽  
...  

Abstract Background: GPIbα is a critically important platelet receptor best recognized for its ability to bind vWF and mediate platelet adhesion. In addition, a GP Ibα binding site for thrombin has been described and characterized for several decades. However, the physiologic relevance of GPIbα/thrombin binding has remained elusive. Site-directed mutagenesis has shown that a sulfated GP Ibα residue, Tyr276, is essential for thrombin binding to GP Ibα but not critical for vWF binding. The importance of Tyr276 was further substantiated in the crystal structure of a GP Ibα/thrombin complex confirming the sulfated Tyr276 GP Ibα residue directly participates with other neighboring negatively charged residues in establishing a first contact with the anion-binding exosite II of an α-thrombin molecule. To determine the physiologic relevance, if any, of the GP Ibα Tyr276 residue we generated mice with platelets containing a mutant human GP Ibα subunit containing a Tyr276 to Phe276 substitution (Y276F) and did comparative studies with mice expressing a normal human (WT) GP Ibα subunit. Methods: Mouse colonies expressing the normal or variant GP Ibα subunits were established by transgenic technology and bred into a mouse colony devoid of murine GP Ibα. Surface expression of WT or Y276F GP Ibα subunits was measured by flow cytometry. Relevant assays included platelet counts, FeCl3-induced thrombus formation, tail bleeding times, hemoglobin content following tail resection (measured as 575 nm absorbance of lysed erythrocytes), and washed platelet aggregation induced by different agonists (fibrillar type I collagen, thrombin, ristocetin, arachidonic acid, and PAR activating peptides). Results: Founder mice were selected with nearly identical levels of surface-expressed WT and Y276F GPIbα subunits. Circulating platelet counts were similar between the 2 colonies. Stirred platelet aggregation assays induced by 0.05, 0.1, 0.5 units/ml thrombin, 10 and 20 mg/ml fibrillar collagen, 1.25 mg/ml ristocetin, 0.25 mM arachidonic acid, 100 mM PAR3 activating peptide and 100 mM PAR4 activating peptide revealed no significant differences between WT and Y276F samples. No statistically significant values were obtained in tail bleeding time assays or hemoglobin loss comparing WT and Y276F animals. However, the time required for a reduction in carotid artery blood flow following FeCl3 injury was significantly prolonged in Y276F animals (10.6±1.6 min vs. 5.8±1.4 min). All WT animals displayed complete occlusion following the initial reduction in blood flow. In contrast, 3 out of 6 Y276F animals did not show complete occlusion and had evidence of fluctuating blood flow suggestive of embolization. Discussion: FeCl3 injury of the mouse carotid artery revealed an essential role for GP Ibα residue Tyr276. In contrast, no differences were seen in tail bleeding time assays or in any platelet aggregation assay. These results are suggestive that thrombin binding to human GPIbα may be relevant for the pathological development of a thrombus. This conclusion is based on both the failure of some carotid arteries from Y276F animals to fully occlude and our observation that all Y276F animals showed an increased length of time for the reduction of blood flow. The results suggest thrombin binding to GPIbα could be significant in terms of enhancing fibrinogen cleavage and therefore stabilizing the development of a platelet-rich thrombus.


Blood ◽  
2006 ◽  
Vol 107 (9) ◽  
pp. 3555-3563 ◽  
Author(s):  
Jaehyung Cho ◽  
Deane F. Mosher

To learn how plasma fibronectin stabilizes platelet-rich thrombi in injured mesenteric arterioles of mice, we studied the impact of plasma fibronectin on platelet thrombus formation ex vivo in a parallel flow chamber. Thrombi were greater on surfaces coated with fibrin cross-linked to fibronectin by activated factor XIII than on surfaces coated with fibrin lacking cross-linked fibronectin or with fibronectin alone. Platelet thrombi were even greater when plasma fibronectin was perfused with platelets, resulting in deposition of the perfused fibronectin in platelet thrombi. The effect of perfused fibronectin on thrombogenesis was lost if fibronectin deposition was blocked by coperfusion with the N-terminal 70-kDa fragment of fibronectin or a peptide based on the functional upstream domain of protein F1 of Streptococcus pyogenes. Increases in thrombus formation were dependent on a platelet activator such as lysophosphatidic acid, amount of fibronectin cross-linked to fibrin, and concentration of fibronectin in the perfusate. The dependency of fibronectin concentration extended into the range of fibronectin concentrations associated with increased risk of coronary artery disease. At such concentrations, the 2 mechanisms for insolubilization of plasma fibronectin—cross-linking to fibrin and assembly by adherent and aggregating platelets—synergize to result in many-fold enhancement of platelet thrombus formation.


1987 ◽  
Author(s):  
T Fujimori ◽  
T Saeki ◽  
K Harada ◽  
M Sato ◽  
N Ohshima

A new agent developed in our laboratory, 4-cyano-5,5-bis(4-methoxyphenyl)-4-pentenoic acid (E-5510), suppressed various human platelet functions in vitro. The compound also showed quite potent ex vivo anti-platelet effects in many experimentalanimals. It is well known that anti-platelet effects are not always parallel to anti-thrombotic effects. Thus, in order to predict the efficacy of E-5510 in thrombotic disorders, the anti-thrombotic effects were examined in 3 different animal models of thrombosis.(1) Anti-thrombotic effect in an extracorporeal shunt model Two hrs after oral administration of the drug to guinea pigs,an extracorporeal shunt between the right carotid artery and the left jugular vein was performed. The thrombus formation on a silk thread inserted in the shunt tube was quantitated by measuring the time from the onset of circulation to the stenosis of blood flow. E-5510 dose-dependently inhibited thrombus formation, the minimum effective dose being 0.03 mg/kg.(2) Effect on microthrombus formation in mesenteric arteriole In order to evaluate the effect on intravascular platelet thrombus formation, thrombosis was induced in vivo in mesenteric arteriole in guinea pigs with filtered light from a mercury lamp and an intravenous fluorescent dye in an intravital microscope system (M. Sato and N. Ohshima, Thromb. Res.,35, 319, 1984). The thrombus formation was quantitated by measuring the time taken for circulating platelets to begin to adhere to vessel wall and the time taken for blood flow to stop completely due to fully developed thrombus. Both the time required for platelet adhesion to vessel wall and for platelet thrombus formation were significantly prolonged after oral administration of E-5510.(3) Effect on pulmonary thromboembolism Acute pulmonary thromboembolism was induced in mice by a bolus intravenous injection of arachidonic acid, and mortality was determined 3 min later. E-5510 dose-dependently reduced pulmonary thromboembolic mortality, and its ED50 was 0.11 mg/kg. The results described above indicate thatE-5510 may have beneficial effects in clinical treatments of thrombotic disease.


Sign in / Sign up

Export Citation Format

Share Document