Ultrasonic Detection of Platelet Aggregation at Variable Shear Rates

1984 ◽  
Vol 14 (6) ◽  
pp. 473-479
Author(s):  
Junji Machi ◽  
Bernard Sigel ◽  
Jose R. Ramos ◽  
Jeffrey R. Justin ◽  
Harold Feinberg ◽  
...  
1992 ◽  
Vol 68 (06) ◽  
pp. 694-700 ◽  
Author(s):  
Roy R Hantgan ◽  
Silvia C Endenburg ◽  
I Cavero ◽  
Gérard Marguerie ◽  
André Uzan ◽  
...  

SummaryWe have employed synthetic peptides with sequences corresponding to the integrin receptor-recognition regions of fibrinogen as inhibitors of platelet aggregation and adhesion to fibrinogen-and fibrin-coated surfaces in flowing whole blood, using a rectangular perfusion chamber at wall shear rates of 300 s–1 and 1,300 s–1. D-RGDW caused substantial inhibition of platelet aggregation and adhesion to fibrinogen and fibrin at both shear rates, although it was least effective at blocking platelet adhesion to fibrin at 300 s–1. RGDS was a weaker inhibitor, and produced a biphasic dose-response curve; SDRG was inactive. HHLGGAK-QAGDV partially inhibited platelet aggregation and adhesion to fibrin(ogen) at both shear rates. These results support the identification of an RGD-specific receptor, most likely the platelet integrin glycoprotein IIb: III a, as the primary receptor responsible for platelet: fibrin(ogen) adhesive interactions under flow conditions, and indicate that platelet adhesion to surface bound fibrin(ogen) is stabilized by multivalent receptor-ligand contacts.


2020 ◽  
Vol 11 (1) ◽  
pp. 20190126 ◽  
Author(s):  
B. J. M. van Rooij ◽  
G. Závodszky ◽  
A. G. Hoekstra ◽  
D. N. Ku

The influence of the flow environment on platelet aggregation is not fully understood in high-shear thrombosis. The objective of this study is to investigate the role of a high shear rate in initial platelet aggregation. The haemodynamic conditions in a microfluidic device are studied using cell-based blood flow simulations. The results are compared with in vitro platelet aggregation experiments performed with porcine whole blood (WB) and platelet-rich-plasma (PRP). We studied whether the cell-depleted layer in combination with high shear and high platelet flux can account for the distribution of platelet aggregates. High platelet fluxes at the wall were found in silico . In WB, the platelet flux was about twice as high as in PRP. Additionally, initial platelet aggregation and occlusion were observed in vitro in the stenotic region. In PRP, the position of the occlusive thrombus was located more downstream than in WB. Furthermore, the shear rates and stresses in cell-based and continuum simulations were studied. We found that a continuum simulation is a good approximation for PRP. For WB, it cannot predict the correct values near the wall.


Blood ◽  
2000 ◽  
Vol 95 (12) ◽  
pp. 3796-3803 ◽  
Author(s):  
Nadine Ajzenberg ◽  
Anne-Sophie Ribba ◽  
Ghassem Rastegar-Lari ◽  
Dominique Meyer ◽  
Dominique Baruch

Abstract The aim was to better understand the function of von Willebrand factor (vWF) A1 domain in shear-induced platelet aggregation (SIPA), at low (200) and high shear rate (4000 seconds-1) generated by a Couette viscometer. We report on 9 fully multimerized recombinant vWFs (rvWFs) expressing type 2M or type 2B von Willebrand disease (vWD) mutations, characterized respectively by a decreased or increased binding of vWF to GPIb in the presence of ristocetin. We expressed 4 type 2M (-G561A, -E596K, -R611H, and -I662F) and 5 type 2B (rvWF-M540MM, -V551F, -V553M, -R578Q, and -L697V). SIPA was strongly impaired in all type 2M rvWFs at 200 and 4000 seconds-1. Decreased aggregation was correlated with ristocetin binding to platelets. In contrast, a distinct effect of botrocetin was observed, since type 2M rvWFs (-G561A, -E596K, and -I662F) were able to bind to platelets to the same extent as wild type rvWF (rvWF-WT). Interestingly, SIPA at 200 and 4000 seconds-1 confirmed the gain-of-function phenotype of the 5 type 2B rvWFs. Our data indicated a consistent increase of SIPA at both low and high shear rates, reaching 95% of total platelets, whereas SIPA did not exceed 40% in the presence of rvWF-WT. Aggregation was completely inhibited by monoclonal antibody 6D1 directed to GPIb, underlining the importance of vWF-GPIb interaction in type 2B rvWF. Impaired SIPA of type 2M rvWF could account for the hemorrhagic syndrome observed in type 2M vWD. Increased SIPA of type 2B rvWF could be responsible for unstable aggregates and explain the fluctuant thrombocytopenia of type 2B vWD.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3197-3197 ◽  
Author(s):  
Yan Yang ◽  
Zhenyin Shi ◽  
Adili Reheman ◽  
Wuxun Jin ◽  
Conglei Li ◽  
...  

Abstract Abstract 3197 Background: Thrombosis and cardiovascular diseases (CVDs) result from blood vessel occlusion by inappropriately activated platelets. They are the leading causes of morbidity and mortality worldwide. Anthocyanins are major phytochemicals abundant in plant food and have been shown to play a protective role against CVDs. Our previous studies have demonstrated that anthocyanins are antioxidative and prevent inflammation (J Biol Chem. 2005; 280:36792-01; Arterioscler Thromb Vasc Biol. 2007; 27:519-24), which may indirectly affect platelet function. It has also been reported that anthocyanins affect platelet activities in whole blood and platelet rich plasma (PRP). However, the direct effects of anthocyanins on platelet function and thrombus formation have not been studied. Methods: Here we investigated the effects of anthocyanins on thrombosis using purified platelets as well as several thrombosis models in vitro and in vivo. Cyaniding-3-gulucoside (Cy-3-g) and delphinidin-3-glucoside (Dp-3-g), the two predominantly bioactive compounds of anthocyanin preparations, were prepared from Polyphenol AS Company in Norway. Purified gel-filtered platelets and PRP from healthy human volunteers and C57BL/6J mice were incubated at 37°C for 10 minutes with different concentrations (0.5μM, 5μM and 50μM) of Cy-3-g, Dp-3-g or PBS buffer as a control. Platelet aggregation was assessed by aggregometry using 5μM ADP, 10μg/ml collagen, or 100μM thrombin receptor activating peptide (TRAP; AYPGKF) as agonists. Platelet adhesion and aggregation were assessed in response to an immobilized collagen matrix in an ex vivo perfusion chamber at both high (1800 s-1) and low (600 s-1) shear rates. The expression of activated GPIIbIIIa was determined via PAC-1 monoclonal antibody in flow cytometry. Lastly, the effects of anthocyanins on thrombus formation in C57BL/6J mice were assessed using a FeCl3-induced intravital microscopy thrombosis model. Results: Both Cy-3-g and Dp-3-g significantly inhibited platelet aggregation induced by collagen and TRAP in gel-filtered platelets, and inhibited aggregation induced by ADP, TRAP and collagen in human and mouse PRP. These inhibitory functions were observed at Cy-3-g and Dp-3-g doses as low as 0.5μM. Cy-3-g and Dp-3-g also reduced the surface expression of activated GPIIbIIIa on resting human platelets in a dose-dependent manner. These compounds also markedly reduced platelet adhesion and aggregation in perfusion chamber assays at both low and high shear rates. Using intravital microscopy, we further demonstrated that Cy-3-g and Dp-3-g decreased platelet deposition, destabilized thrombi, and prolonged the time required for thrombus formation and vessel occlusion. Conclusions: our data clearly demonstrated for the first time that anthocyanin compounds directly inhibited platelet activation, adhesion and aggregation, as well as attenuated thrombus growth at both arterial and veinous shear stresses. These effects on platelets likely contribute to the protective effects of anthocyanins against thrombosis and CVDs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3300-3300
Author(s):  
Reginald Tran ◽  
Byungwook AHN ◽  
David R Myers ◽  
Yongzhi Qiu ◽  
Yumiko Sakurai

Abstract Abstract 3300 Background: Hemostasis is an important physiologic process that requires the aggregation of platelets at distinct sites of vascular injury to promote clot formation and prevent blood loss. Platelet response to soluble agonists and shear stress has been studied extensively, but little is known of how microenvironmental geometry affects platelet function. As platelets must quickly adhere to, aggregate, and initiate coagulation only at the affected areas, spatial cues must at some level regulate this process. This aspect of spatial regulation has been investigated under static conditions by our group and others (Kita et al., 2011; Van de Walle et al., 2012). Understanding this aspect of platelet function is vital for better understanding the process of hemostasis and pathophysiological conditions such as thrombosis. Here, we directly examine how spatial cues affect platelet aggregation and physiology under variable shear conditions by flowing heparinized whole blood over micropatterned collagen in a microfluidic channel. This system allows us to assess platelet aggregate morphology under different geometric constraints and shear rates, as well as evaluate platelet physiology at the single cell level by measuring calcium signaling using fluorogenic dyes. Results: A microfluidic channel was bonded to a glass coverslip stamped with FITC-conjugated Type I collagen using a novel technique combining microcontact printing and the stamp-stick bonding technique (Satyanarayana et al., 2005). Before flowing, each chamber was incubated with 1% bovine serum albumin (BSA) blocking solution for 1.5 hours. Whole heparinized blood was then flowed through the chamber at shear rates of 100, 1000, and 10000 s−1. Platelets were labeled with Fura Red, and time lapse confocal imaging was performed for 10 minutes to monitor the aggregation of platelets at the start of flow. The flow chambers were then flushed with Tyrode's buffer with 0.1% BSA using the same experimental shear rates until the chamber was cleared of red blood cells. Image analysis was conducted using ImageJ (to calculate the percentage of platelet coverage on the collagen stamps at different shear rates. Platelets initially adhere to the distal edge of the collagen micropatterns for all shear rates (Fig. 1), indicating that platelets may require a priming region before forming a stable adhesion. As shear rate increased, platelet coverage of the collagen stamps decreased. However, aggregates also grew without conforming to the geometric constraints imposed by the collagen micropatterns more frequently at those higher shear rates (Fig. 1). Though platelet tethers generally aligned in the direction of the flow, increased tether lengths could be seen when platelets were exposed to higher shear, which may explain why platelets were able to span larger gaps and aggregate in a less spatially constrained manner at high shear rates. Image analysis shows that 51.5% of the collagen was covered by platelet aggregates for a shear rate of 100 s−1 with some platelets forming tethers to span gaps (Fig. 2). When the shear rate was increased to 1000 s−1, platelet coverage of the collagen microstamp drastically dropped to 18.5% (Fig. 2). At a pathophysiological shear rate of 10000 s−1, the percentage of collagen covered by platelets dropped further to 12.8% and adopted a linear shape, although a large portion of the aggregate can be seen spanning gaps between the collagen microstamp (Fig. 1 and 2). Conclusions and Ongoing Efforts: Ours is the first reported study of the spatial regulation of platelet aggregation under variable shear in a microfluidic channel. We have found that platelets are more spatially regulated under low shear conditions compared to high shear, which has implications for thrombosis and other clotting disorders. Future studies will incorporate the simultaneous use of ratiometric fluorgenic calcium signaling dyes to investigate the role of spatial regulation in Ca2+ signaling. Finally, we have developed a method to culture endothelial cells around a collagen micropattern to study this spatial regulation of platelet function under more physiological conditions (Fig. 3). Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 533 ◽  
pp. 119902 ◽  
Author(s):  
Francesco Vetere ◽  
Andrea Mazzeo ◽  
Diego Perugini ◽  
Francois Holtz

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3381-3381 ◽  
Author(s):  
Margo Renee Rollins ◽  
Byungwook Ahn ◽  
Yumiko Sakurai ◽  
Wilbur A Lam

Abstract Introduction: Sickle Cell Disease (SCD) is an inherited monogenic hemoglobin disorder characterized by decreased red blood cells (RBCs) deformability. While RBCs are directly affected by this mutation, the interaction of these cells in the milieu of other components including white blood cells (WBCs), platelets, and soluble factors in whole blood are also thought to contribute to microvascular occlusion in SCD pathophysiology. Several studies have suggested that platelet activation is increased in SCD, but how platelets affect microvascular occlusion is unknown. As cellular interactions are affected by different flow conditions, we leveraged our previous "endothelialized" microfluidic technology (Tsai et al, JCI, 2012) to develop a novel multi-shear microfludic device to investigate blood cell-endothelial cell interactions in 3 different shear rates spanning 3 orders of magnitude ranging from venous to arteriolar shear conditions found in vivo (Figure 1). As platelets are shear-sensitive, this device is conducive to studying platelet interactions in SCD. In addition, we utilized our multi-shear endothelialized microfluidic device for drug discovery, elucidating the mechanism of action of Purified Poloxamer 188 (MST-188). MST-188 is a non-ionic, block copolymer surfactant that has been studied in nearly 400 patients with SCD and is currently being investigated in EPIC (Evaluation of Purified Poloxamer 188 In Crisis), a Phase III trial. MST-188 is composed of a single chain of hydrophobic polyoxypropylene flanked by two hydrophilic polyoxyethylene chains. It is hypothesized to improve microvascular blood flow by reducing viscosity, particularly under low shear conditions, and reducing adhesive frictional forces (Ballas et al 2004). We utilized our endothlelialized multi-shear microfluidic technology to observe cellular interactions in SCD patient samples treated with MST-188. Methods: Whole blood samples were collected from Pediatric patients with HgbSS SCD, including patients on hydroxyurea (HU) via venipuncture in citrate collecting tubes. Samples were recalcified and perfused through a confluently endothelialized multi-shear microfluidic device for 20 minutes. Time-lapse epiflourescence videomicroscopy was obtained to observe cellular interactions under different physiologic flow conditions. Results: Platelet Aggregation in SCD: Using whole blood samples from SCD patients, we observed that platelet aggregation is markedly increased in Hgb SS patients not on HU compared to samples from control and Hgb SS patients on HU (Figure 2). This effect occurs for all shear rates. Attenuation of phosphotadylserine (PS) exposure by MST-188: When a cell undergoes apoptosis, PS "flips" from the intra- to extracellular surface acting as a signal for macrophage engulfment. In order to identify target cell populations a thin smear whole blood from a patient with HgbSS not on HU (Figure 3A). Samples were fluorescently tagged with anti-CD41 to identify platelets and Annexin V to identify the presence of PS (Figure 3B). Patients with HgbSS not on HU have relatively increased fluorescence that is attenuated with treatment with MST-188 (Figure 3C). Conclusion and Future Directions: We have successfully demonstrated a correlation with increased platelet aggregation in endothelialized microfluidic channels in patients with SCD compared to normal controls. The platelets of SCD patients have an increased propensity to aggregate in an abnormal non-shear dependent fashion which correlated directly with fluorescence. This phenomenon appears to be attenuated in patients with SCD on HU in all shear rates. We have also demonstrated that MST-188 attenuates PS exposure mostly found on irreversibly sickled cells. We believe this data and investigational platform to be a good springboard to unravel the utility of targeting platelet specific therapies to augment the course of VOC. This platform can also be used to continue to determine mechanism of action of MST-188 in disease processes, including SCD where inflammation and increased cellular turnover plays a critical role in pathology. Experiments investigating platelet activation markers, co-localization of other cell types including ISCs, reticulocytes and WBC subpopulations with platelet aggregates, as well as characterizing our microfluidic model under de-oxygenated conditions are currently ongoing. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1990 ◽  
Vol 76 (2) ◽  
pp. 345-353 ◽  
Author(s):  
RR Hantgan ◽  
G Hindriks ◽  
RG Taylor ◽  
JJ Sixma ◽  
PG de Groot

We have investigated the molecular basis of thrombus formation by measuring the extent of platelet deposition from flowing whole blood onto fibrin-coated glass coverslips under well-defined shear conditions in a rectangular perfusion chamber. Platelets readily and specifically adhered to fibrin-coated coverslips in 5 minute perfusion experiments done at either low (300 s-1) or high (1,300 s-1) wall shear rates. Scanning electron microscopic examination of fibrin-coated coverslips after perfusions showed surface coverage by a monolayer of adherent, partly spread platelets. Platelet adhesion to fibrin was effectively inhibited by a monoclonal antibody (MoAb) specific for glycoprotein (GP) IIb:IIIa. The dose-response curve for inhibition of adhesion by anti-GPIIb:IIIa at both shear rates paralleled that for inhibition of platelet aggregation. Platelet aggregation and adhesion to fibrin were also blocked by low concentrations of prostacyclin. In contrast, anti- GPIb reduced adhesion by 40% at 300 s-1 and by 70% at 1,300 s-1. A similar pattern of shear rate-dependent, incomplete inhibition resulted with a MoAb specific for the GPIb-recognition region of von Willebrand factor (vWF). Platelets from an individual with severe von Willebrand's disease, whose plasma and platelets contained essentially no vWF, exhibited defective adhesion to fibrin, especially at the higher shear rate. Addition of purified vWF restored adhesion to normal values. These results are consistent with a two-site model for platelet adhesion to fibrin, in which the GPIIb:IIIa complex is the primary receptor, with GPIb:vWF providing a secondary adhesion pathway that is especially important at high wall shear rates.


Sign in / Sign up

Export Citation Format

Share Document