COMPARISON BETWEEN IN VITRO AND IN VIVO AGGREGATION OF PLATELETS

1987 ◽  
Author(s):  
C TAPPARELLI ◽  
P GFELLER ◽  
S SANJAR ◽  
J MORLEY

The extensive utilisation of in vitro tests of platelet aggregation presumes that corresponding effects occur in vivo. Platelet aggregometry in vitro and in vivo have been compared using a range of agonists in order to test this assumption.PRP from citrated peripheral blood of man, rat and guinea-pig was exposed to ADP, adrenaline, serotonin, collagen, thrombin and PAF in a Born aggregometer to define the time course and amplitude of these responses. In rat and guinea-pig, these aggregatory stimuli have also been used to define the time course and amplitude of intrathoracic accumulation of 111 -Indium labelled platelets, using an automated monitoring system (AIMS 8000).Concordance between these tests was evident for ADP, collagen, thrombin and PAF in both species; but, substantial discrepancy was observed between in vitro and in vivo responses to serotonin and adrenaline, since sustained aggregation followed injection of these agonists in the rat.For these platelet stimuli, in vivo aggregometry in rat and guinea-pig may more faithfully reflect the behaviour of human platelets than in vitro studies.

1991 ◽  
Vol 66 (05) ◽  
pp. 609-613 ◽  
Author(s):  
I R MacGregor ◽  
J M Ferguson ◽  
L F McLaughlin ◽  
T Burnouf ◽  
C V Prowse

SummaryA non-stasis canine model of thrombogenicity has been used to evaluate batches of high purity factor IX concentrates from 4 manufacturers and a conventional prothrombin complex concentrate (PCC). Platelets, activated partial thromboplastin time (APTT), fibrinogen, fibrin(ogen) degradation products and fibrinopeptide A (FPA) were monitored before and after infusion of concentrate. Changes in FPA were found to be the most sensitive and reproducible indicator of thrombogenicity after infusion of batches of the PCC at doses of between 60 and 180 IU/kg, with a dose related delayed increase in FPA occurring. Total FPA generated after 100-120 IU/kg of 3 batches of PCC over the 3 h time course was 9-12 times that generated after albumin infusion. In contrast the amounts of FPA generated after 200 IU/kg of the 4 high purity factor IX products were in all cases similar to albumin infusion. It was noted that some batches of high purity concentrates had short NAPTTs indicating that current in vitro tests for potential thrombogenicity may be misleading in predicting the effects of these concentrates in vivo.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2413-2413
Author(s):  
Harika Vemulapalli ◽  
Albayati Samara ◽  
Alexander Y Tsygankov ◽  
Elisabetta Liverani

Abstract Sepsis is a complex clinical syndrome resulting from a serious bloodstream infection. With hospital mortality rates of affected patients reportedly as high as 50%, improved methods for treating sepsis are urgently needed. To begin development of new pharmacologic therapies, we investigated the effect of an antiplatelet treatment on the proliferation of regulatory T cells (Tregs) in a murine model of sepsis. Tregs are a subset of T lymphocytes that downregulate the immune response and promote the resolution of inflammation. Septic patients have elevated levels of circulating Tregs, and this increased prevalence is associated with increased patient mortality. Platelets, which regulate inflammation through cell-cell interactions and through secretion of inflammatory mediators,have been shown to alter the proliferation and activation of Tregs in vitro. However, the influence of platelets on Tregs in vivohas not been fully investigated. We propose that suppression of platelet functions during sepsis may restrain Treg proliferation, leading to the restoration of immunological homeostasis. To study the influence of platelets on Treg proliferation in vivo, we blocked the P2Y12signaling pathway and measured the resulting population sizes of Tregs in septic mice. P2Y12is a Giprotein-coupled purinergic receptor present on platelet surfaces. Stimulation of P2Y12by ADP leads to platelet aggregation and potentiation of platelet secretion. To block the P2Y12signaling pathway, we used the P2Y12antagonist clopidogrel. To induce sepsis in mice, we used cecal ligation and puncture (CLP). Clopidogrelwas administered orallywith a loading dose (30 mg/kg in PBS) one day before surgery and a maintenance dose (10 mg/kg in PBS) two hours prior to surgery. The nonseptic mice in the negative control group (sham) were treated with PBS only. Twenty-four hours after surgery, we isolated cells from the spleens of the mice in each treatment group (sham, CLP, and CLP with clopidogrel) and measured Treg population sizes by incubating the cells with anti-CD4, anti-CD25,and anti-Foxp3 antibodies. Tregs were identified by their positive staining for CD4, CD25, and Foxp3. We found that Tregpopulation sizes were reduced in the septic mice treated with clopidogrel compared with those in the untreated septic mice (Figure 1A).Additionally, we used flow cytometry (forward and side light scattering) to investigatewhether P2Y12antagonism altered the aggregation of platelets and CD4+T cells in whole blood.Platelets and CD4+T cells wereidentified by their positive staining with PE-anti CD41 and FITC-anti CD4, respectively. Events that were double positive for FITC and PE were identified as aggregates and reported as a percentage of gated CD4+T cells.We found that aggregation of platelets and CD4+T cells was reduced in the septic mice treated with clopidogrel (15 ±5 %) compared with that in the untreated septic mice (38 ±6 %) (n= 3, p<0.05 treated CLP vs. untreated CLP). We investigated the effect of blocking the P2Y12signaling pathway in vitrousing co-cultures of human platelets and T cells. Human platelets and T cells were isolated from healthy donors and cultured in the presence or absence of anti-CD3/CD28 (5 μg/mLeach) antibodies for 5 days at 37°C in a humidified atmosphere containing 5% CO2. To block the P2Y12signaling pathway in vitro, we used AR-C69931MX (100 nM). We measured Treg population sizes using flow cytometry as described above. We found that Treg population sizes increased when resting T cells were exposed to platelets, AR-C, or both (Figure 1B). In contrast, we found that Treg population sizes decreased when CD3/CD28-stimulated T cells were exposed to a combination of platelets and AR-C (Figure 1B). Our data indicate that blockade of the P2Y12signaling pathway changes how platelets influence T cells in vitro, depending on whether the T cells have been activated. In conclusion, blockade of the P2Y12signaling pathway restrains Treg proliferation in vivoand in vitro. Our study indicates that targeting platelets to control Treg proliferation and activity may be a promising strategy for treating sepsis. Disclosures No relevant conflicts of interest to declare.


Parasitology ◽  
1983 ◽  
Vol 87 (3) ◽  
pp. 465-479 ◽  
Author(s):  
E. J. Pearce ◽  
Diane J. McLaren

SummaryIn vivoandin vitroparameters of immunity have been assessed in guinea-pigs sensitized with 500 normal or 500 radiation-attenuated cercariae ofSchistosoma mansoni. High levels of resistance to a challenge infection developed in both the chronic and irradiated vaccine model, but immunity was expressed earlier (week 4) and reached higher levels (90%) in the latter case. Vaccinated guinea-pigs have thus been shown to achieve greater resistance than the more commonly used rodent hosts.In vitrocytotoxicity assays have demonstrated that antibodies capable of participating in complement-dependent (lethal antibody) or eosinophil-mediated schistosomular killing, develop in the serum of guinea-pigs immunized with either normal or irradiated cercariae. The time course of development of the eosinophil adherence promoting antibody approximated in both models, the development of immunityin vivo, but the lethal antibody response paralleled the immune status of the animal only in the irradiated vaccine model


1977 ◽  
Author(s):  
M. Blajchman ◽  
A. Senyi ◽  
J. Hirsh

The assessment of the hemostatic function of stored human platelets is difficult to assess in human subjects. The use of thrombocytopenic rabbits treated with ethyl palmitate to produce reticuloendothelial blockade, has made it possible to study the hemostatic function of human platelets in vivo. The assessment of hemostatic function has been made using both a jugular bleeding time technique and an ear bleeding time technique, and in both, a close correlation between bleeding time and platelet count has been established. Using both methods, both fresh and human platelets stored for 72 hours at 22°C correct the bleeding time of thrombocytopenic animals to levels appropriate to the platelet count achieved. Platelets stored at 4°C using standard methods of preparation and storage were ineffective hemostatically after 24 hours storage. Platelets prepared and stored at 4°C at a pH of 6.4 were hemostatically effective in thrombocytopenic rabbits for as long as 10 days of storage. No correlation, however, was noted between the hemostatic effect of stored platelets and in vitro tests of platelet function. Similarly, the intravenous infusion of ADP and collagen produced similar falls in platelet count for both hemostatically effective and non-effective platelets. These studies provide further evidence for the limitations of in vitro tests of platelet function for the assessment of the potential in vivo function of stored human platelets. Furthermore, these findings raise the possibility for the prolongued liquid storage of human platelets at conditions which minimize bacterial contamination, yet maintain hemostatic efficacy.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 582-582
Author(s):  
Monique P. Gelderman-Fuhrmann ◽  
Sheena F. Siddiqui ◽  
Jaroslav G. Vostal

Abstract Evaluation of novel platelet products for transfusion currently involves determination of recovery and survival of radiolabelled platelets in human volunteers since in vitro studies are not predictive of platelet clinical performance and a practical animal model has not been universally accepted. We previously demonstrated that severe combined immunodeficient (SCID) mice could be used as a model to identify severely damaged human platelets (Blood 106 (11), p537a, 2005). To further characterize the sensitivity of this model we used exposure of human platelets to UV light to induce moderate damage, and quantitated this lesion by in vitro tests and in vivo recovery in the SCID mouse model. This type of damage is clinically relevant since UV radiation of blood-platelet concentrates is used to prevent the development of post-transfusion alloimmunization and has been used as part of pathogen reduction systems to inactivate bacteria and viruses. Apheresis platelet products, stored for 1 or 7 days, were exposed to UV light (280–315 nm) for either 20 or 40 min (intensity: 750 μW/cm2). In vitro testing included platelet count, pH, CD62P expression, mitochondrial membrane potential and collagen-induced (2 mg/ml) aggregation. The mitochondrial membrane potential was measured by flow cytometry (FL2-to-FL-1 ratio of platelets loaded with JC-1 (final: 2μM) showed a ratio 6.8 ± 1.5 for the 1 day old platelets and 3.25 ± 0.75 for the 7 day old platelets. Additional flow cytometric testing showed an increase of p-selectin (ant-human CD62P, clone AK-4) expression for 1 day old platelets (31.4 ± 3.5%) and 7 day old platelets (54.6 ± 6.6) after 20 min of UV light. Tables 1 and 2 show in vitro and in vivo results as percent of control (except for pH and JC-1). Table 1. 1 Day Old Platelets UV Exposure Plt Count pH JC-1 Ratio Collagen Aggregation in vivo Recovery 20 min 76.2 7.4 6.5 ± 1.15 101.6 83.8 ± 13.8 40 min 42.2 7.2 3.5 ± 0.4 66.1 56.8 ± 12 Table 2. 7 Day Old Platelets UV Exposure Plt count pH JC-1 Ratio Collagen Aggregation in vivo Recovery 20 min 78.1 7.2 2.85 ± 0.95 116.2 31.8 ± 6.2 40 min 66.1 7.1 nd 5.5 2.27 ± 0.31 For in vivo recovery, approximately 1x10 9 platelets (UV-treated or control) were injected into the tail vein of SCID mice (n=4 per each condition) and serial blood samples were collected. Human platelets were detected in mouse whole blood by flow cytometry using an anti-human GPIIbIIIa mAb (clone P2). Recovery was defined as percent of human platelets in mouse circulation five minutes post infusion. Comparison of recovery between control and UV treatments was done at 4 hours post infusion. UV light treatments, 20 and 40 minutes, reduced the in vivo recovery to 83.8 ± 13.8% and 56.8 ± 12% of control, respectively, for 1 day old platelets and to 31.8 ± 6.2% and 2.27 ± 0.31% of control, respectively, for 7 day old platelets. Our data suggest that the SCID mouse model can identify a relatively subtle UV-induced platelet lesion and that it may be more sensitive than in vitro tests. With further validation, this animal model could be useful for evaluation of the impact of new technologies on the in vivo efficacy of human platelets. The views of the authors represent a scientific opinion and should not be construed as FDA policy.


1987 ◽  
Vol 57 (02) ◽  
pp. 187-190 ◽  
Author(s):  
Dean A Handley ◽  
Ronald G Van Valen ◽  
Christine M Winslow ◽  
John C Tomesch ◽  
Robert N Saunders

SummaryWe have examined a recently developed PAF antagonist SRI 63-675 (dimethyl-tetrahydrofuran-methoxyphosphinyloxy-ethylquinolinium) for its ability to inhibit several major PAF-induced physiological responses. The compound was a potent inhibitor of PAF-induced platelet aggregation in platelet rich plasma obtained from humans, guinea pigs, and rabbits, with IC50 values of 3.43, 0.25, and 0.97 μM, respectively. SRI 63-675 did not inhibit ADP, collagen nor epinephrine-induced human platelet aggregation. The IC50 for inhibition of PAF receptor binding to human platelets was 0.37 μM. In the rat SRI 63-675 inhibited 0.1 μg kg-1 i.v. PAF-induced hypotension, with an ED50 of 32 μg kg-1 i.v. Using the same PAF challenge in the guinea pig, SRI 63-675 inhibited the hemoconcentration (ED50 = 17 μg kg-1 i.v.) and bronchoconstriction (ED50 = 24 μg kg-1 i.v.) responses. In the primate, the ED50 was 28 μg kg-1 i.v. against 3.5 μg kg-1 PAF-induced hemoconcentration. The ratio (1:6) in the primate of PAF used (6.3 nmol kg-1) to antagonist at the ED50 (40.7 nmol kg-1) indicates exceptional potency of SRI 63675 in this species. The inhibition by SRI 63-675 of the major PAF-induced effects in the rat, guinea pig and primate suggests a common receptor may be involved in the expression of these PAF responses.


1979 ◽  
Vol 42 (02) ◽  
pp. 631-640 ◽  
Author(s):  
K A Schumacher ◽  
H G Classen ◽  
M Späth

SummaryIn serum incubated at 36° C for 18-24 hours a factor (DAS) develops which on intravenous injection into cats evokes platelet aggregation followed by an increase in pulmonary vascular resistance (PVR). This change in PVR is mediated via the platelets since it significantly correlates with the preinjection platelet count. There is evidence that phosphatidic acids (PA) and lysophosphatidic acids (LPA) are the active components of DAS. Investigations performed on platelet-rich plasma from man, cat, pig, dog, rabbit, guinea pig, and rat demonstrate that only human and feline platelets exposed to PA or to LPA are aggregated. Feline platelets are more sensitive to either compound than are the platelets from men; however, human platelets exhibit two exceptional properties, a) the sensitivity rapidly declines with time, b) pretreatment with subthreshold concentrations of LPA or PA induces a specific tachyphylaxis.


1972 ◽  
Vol 28 (01) ◽  
pp. 031-048 ◽  
Author(s):  
W. H. E Roschlau ◽  
R Gage

SummaryInhibition of blood platelet aggregation by brinolase (fibrinolytic enzyme from Aspergillus oryzae) has been demonstrated with human platelets in vitro and with dog platelets in vivo and in vitro, using both ADP and collagen as aggregating stimuli. It is suggested that the optimal inhibitory effects of brinolase occur indirectly through the generation of plasma fibrinogen degradation products, without compromising platelet viability, rather than by direct proteolysis of platelet structures.


1980 ◽  
Vol 44 (02) ◽  
pp. 081-086 ◽  
Author(s):  
C V Prowse ◽  
A E Williams

SummaryThe thrombogenic effects of selected factor IX concentrates were evaluated in two rabbit models; the Wessler stasis model and a novel non-stasis model. Concentrates active in either the NAPTT or TGt50 in vitro tests of potential thrombogenicity, or both, caused thrombus formation in the Wessler technique and activation of the coagulation system in the non-stasis model. A concentrate with low activity in both in vitro tests did not have thrombogenic effects in vivo, at the chosen dose. Results in the non-stasis model suggested that the thrombogenic effects of factor IX concentrates may occur by at least two mechanisms. A concentrate prepared from platelet-rich plasma and a pyrogenic concentrate were also tested and found to have no thrombogenic effect in vivo.These studies justify the use of the NAPTT and TGt50 in vitro tests for the screening of factor IX concentrates prior to clinical use.


1993 ◽  
Vol 70 (04) ◽  
pp. 676-680 ◽  
Author(s):  
H F Kotzé ◽  
V van Wyk ◽  
P N Badenhorst ◽  
A du P Heyns ◽  
J P Roodt ◽  
...  

SummaryPlatelets were isolated from blood of baboons and treated with neuraminidase to remove platelet membrane sialic acid, a process which artificially ages the platelets. The platelets were then labelled with 111In and their mean life span, in vivo distribution and sites of Sequestration were measured. The effect of removal of sialic acid on the attachment of immunoglobulin to platelets were investigated and related to the Sequestration of the platelets by the spleen, liver, and bone marrow. Removal of sialic acid by neuraminidase did not affect the aggregation of platelets by agonists in vitro, nor their sites of Sequestration. The removal of 0.51 (median, range 0.01 to 2.10) nmol sialic acid/108 platelets shortened their life span by 75 h (median, range 0 to 132) h (n = 19, p <0.001), and there was an exponential correlation between the shortening of the mean platelet life span and the amount of sialic acid removed. The increase in platelet-associated IgG was 0.112 (median, range 0.007 to 0.309) fg/platelet (n = 25, p <0.001) after 0.79 (median, range 0.00 to 6.70) nmol sialic acid/108 platelets was removed (p <0.001). There was an exponential correlation between the shortening of mean platelet life span after the removal of sialic acid and the increase in platelet-associated IgG. The results suggest that platelet membrane sialic acid influences ageing of circulating platelets, and that the loss of sialic acid may have exposed a senescent cell antigen that binds IgG on the platelet membrane. The antibody-antigen complex may then provide a signal to the macrophages that the platelet is old, and can be phagocytosed and destroyed.


Sign in / Sign up

Export Citation Format

Share Document