Thrombin Generation in Zebrafish

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4218-4218
Author(s):  
Evelien Schurgers ◽  
Martijn Moorlag ◽  
Hilde Kelchtermans ◽  
Coenraad Hemker ◽  
Bas De Laat

Abstract Introduction Recently, the whole blood calibrated automated thrombogram (CAT) was miniaturized enabling the measurement of thrombin generation (TG) in a limited volume (5 µl) of whole blood. Consequently, this approach may be used to determine TG in small lab animals. Zebrafish are readily available test animals for genetic research. However, their small size has been a hurdle in thrombosis and hemostasis research since most assays require large amounts of plasma. In this study we verified the possibility to measure TG in zebrafish using our newly developed miniaturized whole blood assay. Methods For TG, 5 µl of whole blood was mixed with 5 µl of buffer containing a rhodamin-based thrombin-sensitive P2Rho substrate (final concentration (fc) 300 µM). 5 µl of this mixture was put on a paper disk and covered with mineral oil to prevent evaporation. Calibration was done as described previously (Ninivaggi et al. Clin Chem 2012) by adding 5 µl of whole blood to 5 µl of a mixture containing P2Rho (fc 300 µM), a2M-thrombin calibrator (fc 100 nM) and citrate (fc 9,8 mM). Fluorescence was detected with a fluorometer (485/538 nm). Results Due to their limited blood volume, it is impossible to perform both a TG and calibrator measurement on the same fish. Since calibrator measurements performed on blood from different fish demonstrated acceptable variation (CV calibrator slopes < 15%), the average calibrator slope was used for calculations. The calculated TG parameters from 2 independent experiments are depicted in Table 1. TG measured in individual fish showed the same amount of inter-individual variation as in humans. Striking differences with human TG parameters were the short lag time, high peak and high velocity index. Moreover a further analysis of the fibrin network of the clot, by means of scanning electron microscopy (SEM), showed a much denser network composed of thinner fibers compared to humans. Table 1. Thrombin generation parameters and their coefficient of variance (CV) in zebrafish in two independent experiments. Experiment 1(n=4) Experiment 2(n=9) Mean CV Mean CV Peak ETP (nM.min) 575.68 19.69 689.71 24.91 Peak (nM) 1573.16 29.58 1668.05 14.46 Lag time (min) 0.27 0.00 0.49 10.92 Time to peak (min) 1.08 5.30 1.37 11.49 Velocity (nM/min) 4002.33 38.21 3826.35 17.04 Conclusion These results demonstrate the feasibility of measuring TG in whole blood collected from zebrafish. Consequently, zebrafish may be used as a in vivo model to test the effect of (novel) anticoagulant therapeutics on thrombin generation and serve as a model organism for mechanistical research in thrombosis and haemostasis. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5129-5129
Author(s):  
Jawed Fareed ◽  
Debra Hoppensteadt ◽  
Omer Iqbal ◽  
Jeanine M. Walenga ◽  
Bruce E Lewis

Abstract Abstract 5129 Several generic versions of argatroban) (Mitsubishi; Tokyo, Japan) have been introduced in Japan (Argaron, Gartban, Slovastan). In addition, other generic versions of argatroban are being considered by the European and North American regulatory bodies. While the generic versions of argatroban exhibit similar antithrombin potency (Ki values), because of the differential compositional variations their anticoagulant effects in whole blood systems may differ due to their cellular and plasmatic protein interactions. Branded and generic versions of argatroban may exhibit differential anticoagulant actions in the whole blood and plasma based assays due to their differential interactions with blood cells, platelets and plasma proteins. Three generic versions of argatroban that are commercially available in Japan namely Argaron, Gartban and Slovastan and a powdered version of generic argatroban (Lundbeck) were compared with the branded argatroban. Native whole blood thrombelastographic (TEG) analysis was carried out at 0.1 ug/mL, the Activated Clotting Time (ACT) assay was carried out in a concentration range of 0–10 ug/mL, and such coagulation tests as the PT/INR, aPTT, Heptest, and calcium thrombin time were performed. Plasma retrieved from the supplemented whole blood was also assayed. Ratios of the clotting time test values from whole blood and plasma were calculated. Retrieved plasma samples were also assayed in the thrombin generation assays (TGA). All of the different versions of argatroban produced a concentration dependent anticoagulant effect in the native whole blood TEG and ACT. In the TEG, while argatroban and Slovastan showed a similar effect, Gartban, Argaron and a powdered generic showed weaker effects. Argatroban was also different in the ACT assay. At a concentration of 5 ug/ml the ACTs were, Arg 340+15.2 secs, S 297+10.5 secs, G 292.0+19.1 secs and A 285.2+21.7 secs. In the citrated whole blood systems, all agents produced a concentration dependent anticoagulant effect; however, the generic versions produced a stronger anticoagulant effect in comparison to branded argatroban (p<0.001). In the PT assay at 5 ug/mL, argatroban showed 32 ± 3 sec vs 40–50 sec for the generic products. Similarly in the aPTT, Heptest and thrombin time tests argatroban was weaker than the generic products. Differences among generic versions were also evident. Similar results were obtained in the retrieved plasma, however the ratio of whole blood over plasma varied from product to product. The IC50 of the generic and branded argatrobans in the TGA were also different. These results show that while in the thrombin inhibition assays generic and branded argatroban may show similar effects, these agents exhibit assay dependent differences in the whole blood and plasma based assays. Such differences may be more evident in the in vivo studirs where endothelial cells and other interactions may contribute to product individuality. Therefore, based on the in vitro antiprotease assays, generic argatrobans may not be considered equivalent and require a multi-parametric study. Currently available generic argatrobans may not be equivalent in the in vivo anticoagulant effects. Therefore, clinical validation of the clinical equivalence for these drugs is warranted. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 28-28
Author(s):  
Bhavya Doshi ◽  
Courtney Cox ◽  
Bagirath Gangadharan ◽  
Christopher B Doering ◽  
Shannon L. Meeks

Abstract Abstract 28 Hemophilia A is an X-linked recessive disorder that is caused by a deficiency or defect of factor VIII (fVIII) coagulant protein. Approximately 20–30% of patients with severe hemophilia A develop antibodies (Abs) against fVIII (inhibitors) following fVIII replacement therapy, which makes bleeding episodes more difficult to control. Patients with inhibitors are treated with fVIII-bypassing agents such as recombinant factor VIIa (rfVIIa) or activated prothrombin-complex concentrate. However for unknown reasons, some patients display poor hemostatic response to bypass therapy and improved treatment options are needed. Thrombin generation assays provide an in vitro methodology for monitoring bypass therapy in hemophilia (Turecek PL et al. Pathophysiol Haemost Thromb 2003; Varadi K et al. Haemophilia 2004). Recently, it was demonstrated by us and others that combination of fVIII and by-passing agents potentiates in vitro thrombin production in hemophilia A inhibitor plasma (Klintman J et al. Br J Haematol 2010). In our study we investigated the potentiation fVIII confers to fVIIa initiated in vitro thrombin generation using a panel of anti-fVIII Abs with known epitopes. We showed that kinetics of inhibition and Ab epitope were the dominant factors influencing ability of fVIII to potentiate in vitro thrombin production. Specifically, monoclonal Abs targeting only 2 of 11 epitopes, 1 of 3 non-overlapping A2 epitopes and 1 of 2 non-overlapping C2 epitopes, inhibited thrombin generation in a manner that could not be recovered by fVIII supplementation. Here, we analyzed in vitro thrombin generation in epitope-mapped plasmas from 10 patients with hemophilia A and long-standing inhibitors after addition of fVIIa alone or in conjunction with fVIII. Methods: FVIII inhibitor plasmas from 10 patients with hemophilia A were obtained as part of an IRB approved study at the Emory Comprehensive Hemophilia Center. FVIII inhibitor titers and inhibitor kinetics were determined using a modified Bethesda assay. Samples were classified as having type II inhibitors if undiluted plasma resulted in incomplete inhibition of residual fVIII activity (Meeks SL et al. Blood 2007). Thrombin generation assays were carried out in the presence of 2.25 μg/ml recombinant fVIIa in the presence or absence of 1 U/ml recombinant full-length fVIII using reagents purchased from DiaPharma (West Chester, OH). The parameters analyzed include endogenous thrombin potential (area under thrombin generation curve), peak thrombin concentration, time to peak thrombin, lag time (time to 1/6th of peak thrombin) and index velocity (Vi-peak thrombin divided by time to peak minus lag time). Domain specific epitope mapping was carried out using direct ELISA and human/porcine domain hybrid fVIII proteins. Results: Domain mapping of the Abs in the plasmas identified 2 plasmas with predominantly anti-A2 Abs, 4 with predominantly anti-C2 Abs, 2 with both anti-A2 and anti-C2 Abs, and 2 with antibodies that were porcine fVIII cross-reactive (see Table). Plasmas with inhibitor titers less than 25 BU/ml were more responsive to fVIII supplementation with 6 of 7 having increased thrombin generation. Plasmas harboring even trace anti-A2 Abs were more resistant to increased thrombin generation with fVIII supplementation than plasmas with anti-C2 Abs alone. Conclusion: This study suggests a more favorable response to fVIII supplementation of rfVIIa may be predicted by the presence of anti-C2 Abs or inhibitory titers less than 25 BU/ml. In conjunction with our previous monoclonal Ab data, further mapping of epitopes within the fVIII A2 and C2 domains may help improve the ability to predict positive responses to fVIII supplementation of by-passing agents.PatientInhibitor Titer (BU/ml)DomainFVIII InhibitorThrombin Generation (fVIII + fVIIa vs. fVIIa)122A2Type IIIncreased242A2Type IIEqual384C2, small A2Type IEqual47C2Type IIncreased58C2Type IIIncreased620C2Type IEqual78C2Type IIncreased842C2, small A2Type IEqual922Porcine cross-reactiveType IIIncreased105.2Porcine cross-reactiveType IIncreased Disclosures: No relevant conflicts of interest to declare.


2002 ◽  
Vol 87 (02) ◽  
pp. 238-244 ◽  
Author(s):  
J.P. Hérault ◽  
A. Bernat ◽  
C. Gaich ◽  
J.M. Herbert

SummaryThe effect of new heparin mimetics (synthetic oligosaccharides) was studied in vitro with regard to thrombin generation (TG) in rat platelet rich plasma (PRP) and whole blood (WB) and in vivo on stasis-induced venous thrombosis in the rat.TG in PRP and in WB was highly dependent on platelet count and strongly influenced by the haematocrit. The peak of TG appeared to be significantly higher in WB than in PRP whereas the endogenous thrombin potential (ETP) was not significantly different under either condition.The effect of hirudin, the synthetic pentasaccharide SR90107/ Org31540 (SP) and heparin were measured on TG in PRP and WB. We then compared the effect of two new synthetic heparin mimetics (SR121903A and SanOrg123781) with potent and comparable antithrombin (AT) mediated activity against factor Xa and thrombin. These two compounds were made of a pentasaccharide with a high affinity to AT, prolonged at the non-reducing end by an oligosaccharide chain recognised by thrombin. In SR121903A, the charge density and charge distribution was analogous to that of heparin whereas in SanOrg123781 the charges were only located on the last 5 saccharides of the non-reducing end of the molecule. In PRP and in WB, SR121903A acted on the lag time and on the AUC whereas SanOrg123781 inhibited thrombin formation with no effect on the lag time. SanOrg123781 was more potent in inhibiting TG than SR121903A. This difference was due to the structures of the compounds that differed in their ability to be neutralised by platelet factor 4. The antithrombotic effect of the two compounds was examined in a venous thrombosis model in rats. We observed that SanOrg123781 was more active than SR121903A and heparin.Taken together, these results indicate that the activity of oligosaccharides is greatly influenced by the global charge density of the molecule and show that SanOrg123781 is a potent and promising antithrombotic drug candidate.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1216-1216
Author(s):  
Maria Elisa Mancuso ◽  
Veena Chantarangkul ◽  
Armando Tripodi ◽  
Marigrazia Clerici ◽  
Licia Padovan ◽  
...  

Abstract Abstract 1216 Background: The development of inhibitors is the most serious complication of modern hemophilia therapy. Patients with inhibitors are often treated with by-passing agents (namely, aPCC and rFVIIa) whose haemostatic efficacy is not always predictable even in the same subject. Indeed clinical response to by-passing therapies may vary between patients and the lack of a specific laboratory test aimed at monitoring their ability in triggering blood coagulation and at correlating clotting activation to clinical outcome renders the management of these drugs somehow empirical. The thrombin generation (TG) test is a global coagulation assay that may serve as a candidate in this setting. Recently, dose tailoring of by-passing agents was performed using in vitro spiking experiments in order to establish the most adequate doses to cover elective surgery in hemophilic patients with inhibitors. Methods: In this study TG capacity was evaluated for the first time in vivo by drawing plasma samples from hemophilic patients with inhibitors treated either with aPCC or rFVIIa in a non-bleeding state after a minimum wash-out period of 3 days from the last infusion. TG test was performed in platelet-rich (PRP) and platelet-poor (PPP) plasma with the addition of corn trypsin inhibitor (CTI, final concentration 18.3 mcg/ml). Blood was drawn at baseline, 30 minutes, 3 hours (if rFVIIa), 6 hours (if aPCC) and 24 hours after drug administration. Four parameters of the TG curve were evaluated: lagtime, endogenous thrombin potential (ETP), peak and time to peak. Patients were defined as responders to by-passing agents when able to control mild/moderate bleeding episodes by home treatment with those drugs. Results: Eight patients with hemophilia A and high-responding inhibitors (historical peak above 5 BU/ml) with a median age of 33 years (IQR: 20–38) received rFVIIa at a standard dose of 90–120 mcg/kg and aPCC at a dose of 80 IU/kg. In 6 patients the TG test was performed also after the administration of a rFVIIa dose of 270 mcg/kg. Four patients were responders to aPCC, one to rFVIIa, 2 to both drugs and one was non-responder to both. Median values of the TG curve observed at baseline and 30 minutes after drug administration are shown in the Table. Similar variations in the TG curve were observed after administration of either rFVIIa or aPCC or after the administration of rFVIIa at the 2 different dosages. By evaluating only the patients responders to aPCC (n=4), there was no difference in the TG curves obtained after the administration of aPCC or rFVIIa. Conclusions: Our preliminary results show that the in vivo administration of by-passing agents causes an increase of TG capacity in hemophilic patients with inhibitors. However, the extent of such increase does not seem to be related neither with the type nor with the dose of these drugs. The evaluation of TG curve in response to by-passing agents will be performed also in haemophilic patients with inhibitors who underwent or will undergo major surgical procedures. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3646-3646
Author(s):  
Saartje Bloemen ◽  
Marieke de Laat ◽  
Arina ten Cate-Hoek ◽  
Hugo ten Cate ◽  
Bas De Laat ◽  
...  

Abstract Introduction We tested whether the recently introduced measurement of thrombin generation (TG) in whole blood can be used to evaluate the clotting status of patients on vitamin K antagonist (VKA) prophylaxis. The prothrombin time, and hence the International Normalized Ratio (INR), only evaluates the vitamin K dependent factors II, VII and X but not the anticoagulant factors, protein C and S as well as factor IX. In TG all factors play their role and when thrombomodulin (TM) is added the function of proteins C and S is stressed. The thrombotic tendency in congenital protein C resistance proves the importance of this protein C pathway. Aim To compare the INR in samples from patients under VKA prophylaxis to TG in whole blood and in platelet rich and platelet poor plasma (PRP, PPP) both in the presence and in the absence of TM. Materials & Methods Blood samples were collected from 123 consenting patients on VKA. In two thirds (67%) the indication for prophylaxis was atrial fibrillation. Other indications included prosthetic valves, lung embolisms or thrombosis. The INR was determined in the PPP of the samples and the patients were stratified into 5 groups: INR of 1.0 to 1.5, 1.5 to 2.5, 2.5 to 3.5, 3.5 to 4.5 and higher than 4.5. Thrombin generation (TG) was measured via Calibrated Automated Thrombinography (CAT) in whole blood and in PRP and PPP, with and without 20 nM added TM. From the TG curve lag time and time to peak were obtained as well as the maximal thrombin concentration (peak) and the area under the curve (endogenous thrombin potential: ETP). Also red and white blood cells and platelets were counted. Results With increasing INR values, the ETP and peak height decrease and lag time and time to peak prolong. All TG parameters measured in whole blood were significantly correlated (p-values< 0.01) with the values determined in both PRP and PPP. INR was linearly correlated with lag time and time to peak (p-value< 0.01), whereas for the concentration dependent parameters (ETP and peak height) the correlation with the INR was hyperbolical (p-value< 0.01). In plasma, 20 nM TM causes a diminution of ETP and peak of 50-60 % in normals and in patients in the INR 1 – 1.5 group. At higher INR values inhibition is between 25 and 40%, independent of the INR value. In whole blood, on the contrary, the same concentration of TM causes around 30 % of inhibition in normals and in all patients alike. Conclusions Whole blood TG data correlate well with INR and reflect more of the coagulation mechanism than the INR does. Like the INR it does not reflect the function of the VKAs on the natural anticoagulant factors, however. In PPP and PRP addition of TM shows that VKA treatment induces TM resistance in patients with an INR value higher than 1.5. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4989-4989
Author(s):  
Valeria Matus ◽  
José Guillermo Valenzuela ◽  
Claudia G Sáez ◽  
Patricia Hidalgo ◽  
Karla Pereira ◽  
...  

Abstract Acute inflammation in response to severe bacterial infection, results in hemostatic abnormalities ranging from subclinical to sustained systemic clotting activation leading to massive thrombin and fibrin formation and microvascular thrombosis. Endothelial activation and dysfunction are critical determinants of the host response and provide an explanation for the different abnormalities involved in the pathophysiology of sepsis. Infection with pathogenic E. coli may present with a wide spectrum of clinical manifestations, from no symptoms or mild non-bloody diarrhea to severe cases, such as hemolytic uremic syndrome or thrombotic thrombocytopenic purpura, which is characterized by hemolytic anemia and low platelet counts. Although the understanding of the mechanisms that are involved in blood coagulation abnormalities in sepsis has gradually progressed, the role of platelets (Plts) on the procoagulant state during a severe infection remains to be addressed. Human platelets contain functional tissue factor (TF) (Panes et al. 2007) and TFPIa, but it is unknown if bacteria-platelet interaction affects platelet-TF procoagulant or platelet TFPI anticoagulant activities. Moreover, the effects of bacterial activation of platelets on thrombin generation (TG) in platelet rich plasma (PRP) or adhesion to endothelial cells have not been explored. Aims: We assessed the effect of platelet-E. coli interaction on platelet TF-dependent procoagulant activity (PCA), the changes induced by this interaction on platelet TFPI, in TG in PRP and in the adhesive capacity of platelets on cultured HUVEC. Plts activation by E. coli was demonstrated by a significant increase of p-selectin exposure on platelet surface compared to control Plts after 30 min of interaction with this microorganism harvested at exponential phase of growth and incubated in a ratio Plts/bacteria 1:10. Platelet TF-dependent PCA was assessed by FXa generation in washed Plts exposed to E. coli, with addition of exogenous FVIIa and FX, with no extra source of TF. Using the same ratio Plts/bacteria, we observed an increase in FXa after 30 min of incubation, compared with control platelets (p=0.0002, n=12). This enhancement in TF-PCA was concomitant with a decreased expression of TFPI in Plts surface after exposure of PRP to E. coli for 30 min. (p=0.0012, n=9). TG was measured in PRP, previously stimulated by E. coli for 30 min. We observed a shortening in the Lag time and time to peak and a higher thrombin peak in stimulated than in control PRP (p=0.0001, p=0.005 and p=0.0342, respectively, n=12). The reduction in lag time and time to peak was more pronounced than that obtained after eliciting platelet activation with Ristocetin. Preincubation of Plts with E. coli also increased the velocity index of TG compared to PRP alone (p=0,005; n=12). Static adhesion of Pts to endothelium was studied by stimulating fresh washed Plts with E. coli for 30min and then co-incubating them with HUVEC. After 20 min, an increased number of bacterial-activated Plts were adhered to HUVEC, compared with unstimulated Pts. Moreover, visible Plts aggregates were observed, which were positive for fibrin immunostaining, suggesting clot formation during the interaction of Plts with E. coli O111. Our findings show that Plts activated by bacteria results in an enhanced platelet procoagulant activity and adhesion to endothelium. By extension, these in vitro results suggest that platelets play an important role in the prothrombotic state associated with bacterial infections. This work was supported by FONDECYT-Chile 1130835 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2140-2140 ◽  
Author(s):  
P. Collins ◽  
S. Lewis ◽  
N. Macartney ◽  
E. Stephenson ◽  
R. Davies ◽  
...  

Abstract Activation of haemostasis in sepsis may lead to microvascular thrombosis and progression to multiorgan failure (MOF). Almost all critically ill patients with sepsis have abnormal coagulation screens but these are unlikely to adequately represent the state of a patient’s haemostatic system and global assays may be more useful. Normal controls (n=32) and critically ill patients with sepsis (n=39) were recruited. Coagulation factors, antithrombin (AT) and protein C (PC) were measured. Thrombin generation was measured using the calibrated automated thrombogram (CAT) in platelet rich (PRP) and poor (PPP) plasma. Low dose tissue factor (6pM) activated whole blood Rotem® was measured and the first derivative of the trace gave a velocity of clot firmness. Haemostatic changes in sepsis compared to normal controls Controls Controls mean (SD) Sepsis mean (SD) P Apparent effect of change PT (s) 11.7 (0.5) 19.7 (5.9) 0.001 Anticoagulant aPTT (s) 27 (3.4) 44.1 (18.2) 0.001 Anticoagulant Fibrinogen g/l 2.8 (0.57) 5.3 (2.1) 0.001 Prothrombotic FII IU/dl 100 (12.1) 64 (32.2) 0.001 Anticoagulant FV IU/dl 116 (22.9) 96 (55.2) 0.03 Anticoagulant FVII IU/dl 130 (31.1) 58 (33.5) 0.001 Anticoagulant FVIII IU/dl 107 (31.5) 242 (96) 0.001 Prothrombotic FIX IU/dl 101 (16.5) 112 (51.3) NS Neutral FX IU/dl 123 (16.6) 75 (42.1) 0.001 Anticoagulant FXI IU/dl 116 (15.7) 80 (41) 0.001 Anticoagulant FXII IU/dl 125 (27.8) 56 (29.4) 0.001 Neutral PC % 127 (20) 66 (37) 0.001 Prothrombotic AT IU/dl 103 (8) 64 (29) 0.001 Prothrombotic CAT in PRP Lag time (min) 17 (8) 30 (23) 0.02 Delayed ETP (nM.min) 1395 (488) 1270 (573) NS Neutral Peak thrombin (nM) 76 (40) 55 (31) 0.02 Anticoagulant Time to peak (min) 32 (12) 50 (29) 0.001 Delayed CAT in PPP Lag time (min) 2.4 (0.9) 5.1 (5.4) 0.001 Delayed ETP (nM.min) 1681 (281) 1645 (442) NS Neutral Peak thrombin (nM) 454 (100) 343 (146) 0.001 Anticoagulant Time to peak (min) 4.2 (1.2) 6.8 (6.6) 0.001 Delayed Low dose tissue factor Rotem Clot time (s) 818 (271) 1170 (766) 0.04 Delayed Alpha angle (°) 51 (12) 67 (17) 0.005 Prothrombotic MCF (mm) 51 (12) 67 (17) 0.001 Prothrombotic Max vel (mm/s) 6.5 (3.0) 10.9 (7.4) 0.005 Prothrombotic Time to Vmax (s) 1040 (334) 1079 (650) NS Neutral AUC 51 (12) 62 (24) 0.001 Prothrombotic The results show that despite decreased levels of factors II, V, VII, XI and XII (correlation with decreased albumin, P<0.01, suggesting synthetic dysfunction as well as consumption); global measures of haemostasis show delayed but preserved or enhanced overall thrombin generation and clot formation. We hypothesise that the raised FVIII and normal FIX offset the decreased levels of other factors to maintain total thrombin generation which is delayed due to slowed formation of sufficient initial thrombin to activate FV, VIII and XI and stimulate expression of platelet phospholipids. Raised fibrinogen (correlation with CRP, P<0.001), important in the whole blood Rotem-based method, contributes to the prothrombotic state. The global assays are not sensitive to AT and PC and may underestimate the prothrombotic state. These data suggest that haemostatic changes in sepsis are predominantly prothrombotic and may be important in microvascular thrombosis and progression MOF.


Stroke ◽  
2021 ◽  
Vol 52 (Suppl_1) ◽  
Author(s):  
Sarina Falcione ◽  
Gina Sykes ◽  
Joseph Kamtchum Tatuene ◽  
Danielle Munsterman ◽  
Twinkle Joy ◽  
...  

Background and Purpose: Thrombus formation is central to pathophysiology of stroke in patients with atrial fibrillation. Whether factors in plasma contribute to thrombus generation in patients with atrial fibrillation remains unclear. In this study we sought to determine whether plasma contributes to thrombin generation in patients with atrial fibrillation. Methods: There were 78 acute ischemic strokes with atrial fibrillation and 37 non-stroke controls. Plasma thrombin generation was measured by thrombin generation assay, resulting lag time, peak thrombin, time to peak and area under the curve was assessed. Thrombin generation capacity was compared in stroke patients with atrial fibrillation to non-stroke controls. The relationship to anticoagulation was assessed. In vitro, the effect of anticoagulation on plasma thrombin generation was determined. Results: Thrombin generation capacity was increased (shorter lag time and time to peak) in ischemic stroke patients with atrial fibrillation compared to non-stroke atrial-fibrillation controls (p<0.05 and p<0.01, respectively). Anticoagulation decreased plasma induced thrombin generation. Ischemic stroke patients with atrial fibrillation treated with anticoagulation (DOAC or warfarin) had lower plasma induced thrombin generation compared to atrial-fibrillation patients not on anticoagulation (p<0.05). Thrombin generation by plasma could be further reduced by DOAC in an in-vitro assay. Conclusions: Stroke patients with atrial fibrillation have a higher plasma induced thrombin generation compared to atrial fibrillation controls. Factors in plasma such as leukocyte derived tissue factor likely contribute to thrombus formation in patients with atrial fibrillation. As such, components in plasma may represent new targets to reduce thrombus formation and stroke risk in patients with atrial fibrillation.


2017 ◽  
Vol 6 (2) ◽  
pp. 89-99 ◽  
Author(s):  
Malin Nylander ◽  
Signe Frøssing ◽  
Caroline Kistorp ◽  
Jens Faber ◽  
Sven O Skouby

Polycystic ovary syndrome (PCOS) is associated with increased risk of venous thromboembolism (VTE) and cardiovascular disease (CVD) in later life. We aimed to study the effect of liraglutide intervention on markers of VTE and CVD risk, in PCOS. In a double-blind, placebo-controlled, randomized trial, 72 overweight and/or insulin-resistant women with PCOS were randomized, in a 2:1 ratio, to liraglutide or placebo 1.8 mg/day. Endpoints included between-group difference in change (baseline to follow-up) in plasminogen activator inhibitor-1 levels and in thrombin generation test parameters: endogenous thrombin potential, peak thrombin concentration, lag time and time to peak. Mean weight loss was 5.2 kg (95% CI 3.0–7.5 kg, P < 0.001) in the liraglutide group compared with placebo. We detected no effect on endogenous thrombin potential in either group. In the liraglutide group, peak thrombin concentration decreased by 16.71 nmol/L (95% CI 2.32–31.11, P < 0.05) and lag time and time to peak increased by 0.13 min (95% CI 0.01–0.25, P < 0.05) and 0.38 min (95% CI 0.09–0.68, P < 0.05), respectively, but there were no between-group differences. There was a trend toward 12% (95% CI 0–23, P = 0.05) decreased plasminogen activator inhibitor-1 in the liraglutide group, and there was a trend toward 16% (95% CI −4 to 32, P = 0.10) reduction, compared with placebo. In overweight women with PCOS, liraglutide intervention caused an approximate 5% weight loss. In addition, liraglutide affected thrombin generation, although not significantly differently from placebo. A concomitant trend toward improved fibrinolysis indicates a possible reduction of the baseline thrombogenic potential. The findings point toward beneficial effects of liraglutide on markers of VTE and CVD risk, which should be further pursued in larger studies.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4029-4029
Author(s):  
Wolfgang Wegert ◽  
Manuela Krause ◽  
Inge Scharrer ◽  
Ulla Stumpf ◽  
Andreas Kurth ◽  
...  

Abstract The changes of tissue factor (TF) blood levels in patients undergoing major surgery has been reported presenting controversial data. Whether this TF is hemostatically active or if it interacts with other coagulation factors, e.g. FVIII, is still unclear, making thrombotic risk and complications assessment for even more difficult. We analyzed plasma samples from four male patients aged 27–55 with severe hemophilia A without inhibitory antibodies, undergoing total knee replacement, which all gave informed consent. Initial FVIII doses before intervention was 75–80 U/kg. Treatment following intervention was targeted at 100 % FVIII serum levels. None received heparin. No bleeding events occurred during the observation period. The samples were taken at these timepoints (TP): 1. before preoperative FVIII substitution, 2. at the time of first incision (intervention start), 3. at circulation arrest release + 90 s after prosthesis implantation, 4. final suture (intervention end), 5. 24 h and 6. 48 h after intervention to assay procedurally induced TF production. Coagulation analyses were carried out using a fluorometric thrombin generation assay (TGA) in platelet rich plasma (PRP), RoTEG (rotation thrombelastography) in whole blood and a TF ELISA for the plasma samples’ TF levels. Both clotting function tests were started using TF diluted 1:100.000 and calcium chloride 16,7 mM (final conc.). TGA parameters were ETP, PEAK (maximum thrombin generation velocity), TIME TO PEAK, LAG TIME. TGA parameters directly related to thrombin activity (ETP; PEAK) showed no change during the intervention, but a sharp decrease 24 h later with a partial recovery 48 h later. TGA time marks (TIME TO PEAK, LAG TIME) changed in an inverse way, except for the difference from LAG TIME and TIME TO PEAK, which shortened continously after circulation arrest removal. RoTEG was characterized using 4 parameters: clotting time (CT), clot formation time (CFT), maximum clot firmness (MCF) and clot formation rate (CFR). After preoperative FVIII substitution, CT decreased by 10 % and CFT by 50 % in 48 h. MCF stayed unchanged during the intervention and the following 24 h, but increased by 20 % at 48 h. CFR increased by 10 % during intervention, and by 20 % from 24 to 48 h. TF ELISA showed preoperative TF plasma levels from 11 to 271 pg/ml. After release of circulation arrest (TP 3) TF concentration increased sharply (4 times the initial value), which was not detectable in the samples taken at TPs 2 and 4. TF levels further increased at TPs 5 and 6 to 170 % and 317 % resp. Altogether, TF plasma levels elevated after major surgery seem to correspond to a potential risk factor for postoperative thrombosis, especially when elevation is induced after intervention. However, functional coagulation assays do not change uniformly, as the thrombin generation assay reflects no marked changes under intervention, but in the period after(24–48 h). Changes in the RoTEG whole blood clotting assay are not dramatic but indicate a thrombophilic shift in coagulation balance also pronouned at 24–48h, too. These results demonstrate that increased coagulability after orthopedic surgery detected using functional clotting assays correlates with increased TF levels, but further studies must be performed to prove this relation in healthy individuals.


Sign in / Sign up

Export Citation Format

Share Document