scholarly journals Insights into Endogenous Vs Exogenous Cargo-Containing Platelet Alpha-Granules

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1028-1028
Author(s):  
Sergey Zaytsev ◽  
Hyunsook Ahn ◽  
Victoria Stepanova ◽  
M. Anna Kowalska ◽  
Khalil Bdeir ◽  
...  

Abstract Alpha granules in megakaryocytes contain a mixture of endogenously expressed proteins as well as proteins taken up from the intramedullar fluid. Both pools are thought to be found in all alpha granules in the megakaryocytes and released platelets. We have been studying the ectopic expression of urokinase (uPA) in platelets as a targeting strategy for fibrinolysis of nascent thrombi without causing fibrinolysis of established thrombi. These studies also demonstrated that there are two distinct pools of alpha granules, an endogenous cargo pool of granules and an exogenous uptake cargo pool of granules. Using in vitro grown megakaryocytes from two sources (1) CD34+-hematopoietic progenitor cells and (2) induced-pluripotent stem cell derived line imMKCL kindly provided by Dr. Koji Eto at Kyoto University, we demonstrated that urokinase can be localized within alpha granules in the megakaryocytes by either adding urokinase to the media or by ectopically expressing the protein using a lentiviral strategy. We observed that both a human single-chain uPA (scuPA) or a plasmin-insensitive but thrombin-activatable truncated human uPA mutant (uPA-T) in the media were internalized into granules distinct from granules containing ectopically expressed mouse scuPA following lentiviral transduction. Endocytosed uPA showed no co-localization with endogenous von Willebrand Factor (vWF), but significant colocalization with endocytosed Factor V or plasminogen (PLG) on confocal immunofluorescent microscopy. Further, Factor V competed with both uPA variants for uptake from the media. Uptake of these proteins was inhibited by the LRP1 antagonist receptor-associated protein (RAP) and by anti-LRP1 antibodies. This suggests that both proteins use the same endocytic receptor pathway and share this pathway with other proteins taken up from the media, including Factor V. We found that in vitro-generated CD34+ megakaryocytes pre-loaded with exogenously added PLG and co-incubated thereafter with recombinant scuPA and FV significantly degraded FV; however, no vWF degradation was observed in CD34+-derived megakaryocytes that had endocytosed or ectopically expressed scuPA with exogenously added PLG, suggesting that only the proteins which are endocytosed by in vitro-generated megakaryocytes are degraded by uPA-generated plasmin, whereas endogenous alpha-granular proteins remain intact. We then asked whether uPA localized in these two distinct pools can be released at sites of nascent thrombus formation and be effective in preventing nascent thrombus growth. We infused CD34+-derived MKs into NOD-scid IL2rγnull (NSG) mice homozygous for VWF R1326H (a mutation switching binding VWF specificity from mouse to human GPIb/IX). NSG/VWF R1326H mice have impaired clotting after vascular injury compared to NSG mice unless infused with human platelets or MKs . Significantly less post-injury clotting was seen upon infusion of either endogenous or exogenous scuPA-containing MK infusion. Further studies to define relative efficacy at the same levels of scuPA are being pursued. These studies show that there are two sets of alpha granules that remain separate during megakaryopoiesis in vitro: granules with endogenously expressed cargo and granules with endocytosed cargo with limited mixing between the two pools by confocal microscopy studies and following PLG uptake studies. The extent of mixing that occurs subsequently in released platelets was not studied nor has these finding been done with primary MKs not grown in culture; however, we believe that these studies extend our understanding of the nature of alpha granules and offer new insights into how to manipulate their cargo. Disclosures Cines: Dova: Consultancy; Rigel: Consultancy; Treeline: Consultancy; Arch Oncol: Consultancy; Jannsen: Consultancy; Taventa: Consultancy; Principia: Other: Data Safety Monitoring Board.

2014 ◽  
Vol 115 (suppl_1) ◽  
Author(s):  
Suresh K Verma ◽  
Prasanna Krishnamurthy ◽  
Alexander R Mackie ◽  
Erin E Vaughan ◽  
Mohsin Khan ◽  
...  

The association of inflammation with atherosclerosis and restenosis is now fairly well established. Restenosis, a persistent complication of percutaneous vascular interventions, is thought to be a complex response to injury, which includes early thrombus formation, neointimal growth and acute inflammation. Mononuclear phagocytes are likely participants in the host response to vascular injury, via the secretion of cytokines and chemokines, including TNF-alpha (TNF). Others and we have previously shown that IL-10 inhibits TNF and other inflammatory mediators produced in response to cardiovascular injuries. The specific effect of IL-10 on endothelial cell (EC) biology is not well elucidated. Here we report that in a mouse model of carotid denudation, IL-10 knock-out mice (IL10KO) displayed significantly delayed ReEndothelialization and enhanced neointimal growth compared to their WT counterparts. Exogenous treatment of recombinant IL-10 dramatically blunted the inflammatory cell infiltration and neointimal thickening while significantly accelerating the recovery of the injured endothelium both WT and IL10KO mice. In vitro, IL10 co-treatment reversed TNF-mediated growth arrest, EC cell cycle inhibition, EC-monocyte adhesion and EC apoptosis. At signaling level, IL-10 reduced TNF-induced activation of JNK MAP kinase while simultaneously activating PI3K/Akt pathway. Because IL-10 function and signaling are important components for control of inflammatory responses, these results may provide insights necessary to develop strategies for modulating vascular repair and other accelerated arteriopathies, including transplant vasculopathy and vein graft hyperplasia.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 513-513
Author(s):  
Kyoichi Ogata ◽  
Steven W. Pipe

Abstract Hemophilia A results from the quantitative or qualitative deficiency of coagulation factor VIII (FVIII). FVIII is synthesized as a single-chain polypeptide of approximately 280 kDa with the domain structure A1-A2-B-A3-C1-C2. Whereas the A and C domains exhibit ~40% amino acid identity to each other and to the A and C domains of coagulation factor V, the B domain is not homologous to any known protein and is dispensable for FVIII cofactor activity. Missense mutations in the FVIII B domain have been described in patients with variable phenotypes of hemophilia A. According to the NCBI SNPs (single nucleotide polymorphism) database, 22 SNPs are reported within FVIII, 11 of which occur within the B domain. FVIII B domain variant D1241E has been reported as a missense mutation associated with mild or severe hemophilia A, yet this mutation is also present in the NCBI SNPs database. We hypothesize that D1241E and most other reported B domain missense mutations are not the causative mutation for hemophilia A in these patients but represent SNPs or otherwise non-pathologic mutations. To investigate this, we analyzed 7 B domain missense mutations that were previously found in hemophilia A patients (T751S, V993L, H1047Y, D1241E, T1353A, P1641L and S1669L). Comparative analysis showed that the amino acids at these positions are not conserved in all species and in some cases, the amino acid substitution reported in hemophilia patients is represented in the native sequence in other species. Analysis with PolyPhen Software showed that only H1047Y mutation was considered as “possibly damaging”, while the others were considered as “benign”. To investigate this further, we constructed seven plasmid vectors containing these B domain missense mutations. The synthesis and secretion of FVIII wild-type (WT) and these seven mutants were compared after transient DNA transfection into COS-1 monkey cells in vitro. Analysis of the FVIII clotting activity and antigen levels in the conditioned medium demonstrated that all mutants had FVIII activity and antigen levels similar to FVIII WT. Further, FVIII WT, H1047Y and D1241E mutants were introduced into a FVIII exon 16 knock-out mouse model of hemophilia A by hydrodynamic tailvein injection in vivo. The mouse plasma was analyzed at 24 hrs for activity and antigen expression. Mutants H1047Y and D1241E expressed at 211 mU/mL and 224 mU/mL activity with FVIII antigen levels of 97 ng/mL and 118 ng/mL, respectively, similar to FVIII WT. These results suggested that H1047Y and D1241E mutants did not lead to impairments in secretion or functional activity. We conclude that most missense mutations within the FVIII B domain would be unlikely to lead to severe hemophilia A and that the majority of such missense mutations represent polymorphisms or non-pathologic mutations. Investigators should search for additional potentially causative mutations elsewhere within the FVIII gene when B domain missense mutations are identified.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 2524-2524
Author(s):  
Suman Paul ◽  
Jacqueline Douglass ◽  
Annika Schaefer ◽  
Emily Han-Chung Hsiue ◽  
Alexander Pearlman ◽  
...  

2524 Background: Increased tumor suppressor protein p53 expression is observed in a wide range of human cancers. As a result there is intense interest in targeting p53 for cancer therapy. Intracellular p53 is inaccessible to therapeutic antibodies that bind cell surface proteins. However, intracellular proteins including p53 are degraded into peptides that are presented on cell surface in association with HLA class I molecules. Thus p53 peptide-HLA (p53-HLA) complexes can be antibody targets. Methods: Using phage display we identified a novel anti-p53-HLA single chain variable fragment (scFv) clone-43 that recognizes a wild-type p53 10-mer epitope bound to HLA-A*2402. By coupling our clone-43 scFv with an anti-CD3 scFv, we generated a single chain diabody (scDb) designed to activate T-cells against p53-expressing target cells. Results: In-vitro co-culture of clone-43 scDb with donor human T-cells and p53 expressing SIG-M5 cancer cells results in SIG-M5 cell killing and concomitant T-cell interferon gamma (IFNγ) release. In contrast, similar co-culture with SIG-M5 p53-knock out (KO) cells showed no cell killing and minimal IFNγ release demonstrating specificity of clone-43 to p53 expressing cells. Additionally, in-vivo growth of p53 expressing SW480 cancer cell xenografts in NSG mice was completely terminated by clone-43 scDb injections. A major concern for wild-type p53 epitope targeting is potential on-target off-tumor effect on non-cancerous tissue. We observed significant in-vitro clone-43 scDb mediated killing of human HLA-A*24:02 peripheral blood mononuclear cells. To better evaluate effect of clone-43 scDb on non-neoplastic human cells, we engrafted HLA-A*24:02 human CD34+ hematopoietic stem cells into NSG mice to generate a humanized mouse model with circulating mature human CD45+ cells. Clone-43 scDb treatment resulted in selective depletion of circulating human cells while the same cells persisted in mice treated with unrelated control scDb. Conclusions: Our observation that immune targeting of wild-type p53 epitope results in significant off-tumor hematopoietic cell death is contrary to previously published reports and carries important implications for future anti-p53 antibody and vaccine design for cancer immunotherapy.


2019 ◽  
Author(s):  
Jin Wook Hwang ◽  
Christophe Desterke ◽  
Olivier Féraud ◽  
Stephane Richard ◽  
Sophie Ferlicot ◽  
...  

SUMMARYHereditary cancers with cancer-predisposing mutations represent unique models of human oncogenesis as a driving oncogenic event is present in germline, exposing the healthy member of a family to the occurrence of cancer. The study of the secondary events in a tissue-specific manner is now possible by the induced pluripotent stem cell (iPSC) technology offering the possibility to generate an unlimited source of cells that can be induced to differentiate towards a tissue at risk of malignant transformation. We report here for the first time, the generation of a c-met-mutated iPSC lines from the somatic cells of a patient with type 1 papillary renal cell carcinoma (PRCC). We demonstrate the feasibility of kidney differentiation with iPSC-derived organoids expressing markers of kidney progenitors with presence of tight junctions and brush borders in tubular structures at transmission electron microscopy. Importantly, c-met-mutated kidney organoids expressed PRCC markers both in vitro and in vivo in NSG mice. Gene expression profiling of c-met-mutated iPSC-derived organoid structures showed striking molecular similarities with signatures found in a large cohort of PRCC patient samples and identified 11 common genes. Among these, BHLHE40 and KDM4C, well-known factors involved in PRCC pathogenesis, were expressed in c-met-mutated kidney organoids. This analysis applied to primary cancers with and without c-met mutation showed overexpression of the BHLHE40 and KDM4C only in the c-met-mutated PRCC tumors, as predicted by c-met-mutated organoid transcriptome. These data represent therefore the first proof of concept of the generation of “renal carcinoma in a dish” model using c-met-mutated iPSC-derived organoids, opening new perspectives for discovery of novel potentially predictive disease markers and novel drugs for future precision medicine strategies.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5460-5460
Author(s):  
Noriko Konishi ◽  
Katsuhiko Hiroe ◽  
Yasuhiro Imaeda ◽  
Takuya Fujimoto ◽  
Keiji Kubo ◽  
...  

Abstract Thrombin generation serves to amplify the coagulation cascade via positive feedback activation of factor V (FV) and factor VIII. We hypothesized that factor Xa (FXa) inhibitors, unlike thrombin inhibitors, would not block the feedback activation of the coagulation cascade but would have a favorable anticoagulating profile—sufficient to prevent thrombus formation, yet not interfere with hemostatic plug formation. TAK-442 is a newly synthesized, selective FXa inhibitor that strongly inhibits FXa (with a Ki value of 1.8 nM), and displays more than 440x selectivity toward FXa than other serine proteases. In the present study, we compared the effects of TAK-442 versus ximelagatran on FV-mediated positive feedback in vitro, and on their antithrombotic and hemorrhagic effects in a rat model of venous thrombosis. In vitro, TAK-442 gradually inhibited thrombin generation and prolonged prothrombin time (PT) in a dose-dependent manner, while melagatran, an active form of ximelagatran, exhibited a steeper effect at higher doses tested. The PT prolonging potency was increased in FV–deficient human plasma, with CT2 values (the concentration that causes 2 times prolongation of clotting times) of 120 nM for TAK-442 and 32 nM for melagatran, compared with 500 nM and 360 nM for TAK-442 and melagatran, respectively in normal plasma. In the rat model of venous thrombosis, TAK-442 (10 mg/kg, po) prevented thrombus formation by 55% and prolonged PT by 1.3 times of control values; a similar effect was observed in ximelagatran-treated (3 mg/kg, po) animals, with 59% inhibition of thrombus formation and 1.2 times prolongation of PT. TAK-442 at 100 mg/kg, prolonged PT by 2.1 times, with no significant change in bleeding time (BT); in contrast, increasing the dose of ximelagatran to 10 mg/kg, po prolonged PT by 3.9 times and significantly (P<0.025) increased BT. Our data suggest that the differential effects of the two agents on FV-mediated amplification of thrombin generation may underlie the observation of a wider therapeutic window for TAK-442 than for ximelagatran.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 896-896
Author(s):  
Karen Silence ◽  
Heidi Jonckheere ◽  
Peter Casteels ◽  
Jan Roodt ◽  
Muriel Meiring ◽  
...  

Abstract In patients with plaque rupture, platelets adhere, aggregate and form a thrombus. Current strategies to prevent thrombus formation consist of the use of Aspirin®, Plavix® and integrin αIIbβ3 blockers (e.g. Reopro®) in combination with Heparin®. These drugs are associated with high bleeding risk. Several in vivo experiments have shown that neutralizing the collagen von Willebrand Factor (vWF) platelet glycoprotein (GP)Ib-IX-V axis strongly inhibits arterial thrombosis without bleeding complications, therefore, these targets are of high interest to develop new anti-thrombotic drugs. Nanobodies are antibody-derived therapeutic proteins with the structural and functional properties of naturally occurring single-chain antibodies derived from camelids. ALX-0081 is a bivalent humanized Nanobody targeting the GPIb-IX-V binding site at the A1 domain of vWF. The precursor molecule was isolated from a llama immunized with the recombinant A1 domain of vWF and then humanized and engineered into a bivalent format to maximally benefit from the avid binding to vWF. In vitro, ALX-0081 can completely inhibit platelet adhesion to collagen at nanomolar concentrations. This inhibition is specific for the high shear rates relevant for coronary and carotid arteries whilst platelet adhesion and aggregation under low shear conditions is unaltered. The Nanobody also inhibits platelet adhesion to ultra large vWF (ULvWF) whilst it does not inhibit cleavage of ULvWF by ADAMTS-13. In a modified Folt’s model in baboons ALX-0081 inhibits thrombus formation more efficiently than a combination of Aspirin, Heparin and Plavix. Inhibition of thrombus formation is sustained in the presence of epinephrine and upon a new injury confirming the strong anti-thrombotic effect of ALX-0081. The Nanobody is effective at doses approximately 10–20 times lower than the dose required for Reopro. Ex vivo analysis of plasma samples after ALX-0081 administration in baboons in the ristocetin induced platelet aggregation (RIPA) assay reflects the efficacy seen in the Folt’s model. Therefore, this assay seems to be suited to predict effective ALX-0081 concentrations in vitro. In comparison to Reopro and Plavix, ALX-0081 is associated with less bleeding complications, even at doses exceeding the effective dose by a factor of 10 probably because of its selective inhibition of platelet aggregation under high shear but not under low shear conditions. After treatment with ALX-0081 no effect on other hematological parameters such as PT, aPTT, platelet count, VWF concentration and FVIII levels is seen and no immunogenicity is detected in baboons after repeated administration of ALX-0081. The terminal half-life of ALX-0081 in baboons is 8 hours, indicating that the molecule adopts the half-life of vWF. This high efficacy combined with an improved safety compared to the currently marketed drugs suggests that Ablynx’ drug development candidate ALX-0081 can become a powerful drug to treat acute thrombotic events in indications such as ACS, stroke, and TTP.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1837-1837
Author(s):  
A. Valance Washington ◽  
Sébastien Gibot ◽  
Ismael Acevedo ◽  
Alina De La Mota ◽  
James Gattis ◽  
...  

Abstract TREM-like transcript-1 (TLT-1) is a novel, platelet-specific, membrane protein that is stored in a-granules and brought to the platelet surface upon activation. Little is known about its function. Here we build upon our recent findings that a soluble fragment of TLT-1 (sTLT) is released upon platelet activation and can be detected in the serum, but not the plasma, of healthy individuals. Evaluation of patients admitted to the University Hospital, Ramon Ruiz Arnau, in Bayamon Puerto Rico with the diagnosis of sepsis for the presence of plasma sTLT demonstrated significant levels of sTLT in septic individuals as compared to controls, indicating that TLT-1 plays a role in the early stages of sepsis. To directly assess this possibility we generated TLT-1 knockout mice and characterized their hemostatic function. Compared to wild type mice, TLT-1−/− platelets displayed decreased aggregation when treated with thrombin, collagen, ADP, or U46619 in vitro. The reduced platelet aggregation of TLT-1−/− mice translates to a 30% increase in bleeding time in tail snip assays compared to controls, supporting TLT-1’s role in vascular homeostasis. To evaluate TLT-1’s role in sepsis we challenged mice with lipopolysaccharide (LPS). LPS challenged TLT-1 null mice die faster and have a significantly lower rate of survival than wild type mice. We used the localized Shwartzman reaction to model the pathology associated with sepsis in a controlled lesion. Although neutrophil infiltration, thrombus formation, and the formation of microclots was not significantly higher in TLT-1−/− mice, the hemorrhage associated with Shwartzman lesions of these mice was nearly double and lesion size was greater than three times that of wild type mice. Moreover, we demonstrate a recombinant sTLT augments platelet aggregation in vitro suggesting a mechanistic explanation for the increased hemorrhage in TLT-1−/− mice. Thus these studies reveal an important role for TLT-1 in the maintenance of vascular hemostasis by facilitating platelet aggregation through incorporation of sTLT into the forming clot. This work suggests that therapeutic intervention using tools such as anti-TLT-1 single chain antibodies that inhibit platelet aggregation may provide a novel approach to the control of the thrombotic response.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 693-693
Author(s):  
Alexander Schlachterman ◽  
Jianhua Liu ◽  
Yi-Lin Liu ◽  
Katherine High ◽  
Valder Arruda

Abstract The amelioration of hemophilia phenotype or delayed onset of the bleeding episodes in subjects with severe hemophilia A (FVIII deficiency) has been associated with inherited resistance to activated protein C due to factor V Leiden (R506Q), FVL. These observations were confirmed by in vitro systems in which homozygous phenotype of FVL increased thrombin formation in presence of < 1% FVIII by nearly 5-fold (Blood 90:3067). Here we provide in vivo evidence of the beneficial interaction of FVL on the hemophilia phenotype in mice. Animals with severe deficiency of factor VIII due to deletion of intron 16 of factor VIII gene (HA) or large gene deletion of factor IX (HB) were crossed with FVL homozygous mice [+/+] on C57Bl6 strain [Cui et al. (Blood 96:4222)] We used a modified activated partial thromboplastin time (aPTT) assay to compared clotting times among male HA (n=30), HA/FVL [+/+] (n=7), FVL [+/+] (n=5), and litermate WT mice (n=6) with age ranging from 6–12 weeks. Blood samples were collected by tail vein transection into 3.8% sodium citrate. Values for the aPTT in male HA were 67.3 ± 4 sec, and among WT or FVL [+/+] values were 37 ± 4 sec or 31± 2 sec, respectively. Whereas intermediate aPTT values of 53 ± 3.7 sec were determined in HA/FVL [+/+], which differs from HA mice (p<0.0001) but also from WT or FVL (p<0.0001). A similar shortening of aPTT was also determined among HB/FVL[+/+] which were compared to HB, 55 ± 7 sec vs. 64 ± 0.9. Hemostatic challenge by tail clipping assay failed to revealed differences in bleeding times/blood loss among hemophilia animals with or without FVL mutation. To test whether a more sensitive technique would provide further evidence of the improved hemostasis in HA/FVL mice, we assessed real-time in vivo thrombus formation by confocal and widefield microscopy. Mice were anesthetized and the cremaster muscle was exposed for intravital microscopy. Infusion of fluorescently labeled antibody to murine platelet glycoprotein IIb/IIIa complex via the jugular vein allowed monitoring of platelet deposition upon laser-mediated endothelial injury at several sites of the arterial vessel wall. No thrombus formation was observed in severe HA mice following successive vascular injuries, a finding also common in severe HB mice. However, infusion of factor VIII concentrated clearly induced the thrombi formation upon vascular injury. HA/FVL mice tested presented thrombus formation in a comparable fashion of HA-FVIII transfused mice. These in vivo data provide support to the hypothesis that the FVL mutation has the potential to improve the phenotype of severe hemophilia and may offer a novel therapeutic target for hemophilia.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2628-2628
Author(s):  
Jack D Stopa ◽  
Donna S. Neuberg ◽  
Maneka Puligandla ◽  
Bruce Furie ◽  
Robert C. Flaumenhaft ◽  
...  

Abstract Protein disulfide isomerase (PDI) is a ubiquitously expressed oxidoreductase that serves an essential role in protein folding in the endoplasmic reticulum by reshuffling disulfide bonds within nascent proteins. PDI can be released from vascular cells, including platelets, and inhibition or platelet-specific deletion of PDI blocks thrombus formation in vivo. However, the specific function of PDI in thrombus formation is poorly understood. Unlike the role of proteases in blood coagulation, which have been studied in depth, little is known about PDI substrates in the vasculature. Several platelet and endothelial integrins have been identified as putative substrates for PDI, but whether coagulation factors are directly targeted by extracellular PDI has not been established. We now identify platelet factor V as a principal coagulation substrate of extracellular PDI. We developed an unbiased strategy to identify novel substrates of PDI in washed platelets using PDI variants capable of trapping substrates: FLAG-tagged PDI mutants modified by a substitution of arginine or proline for histidine (CGHC → CGRC; CGHC → CGPC) in the catalytic motif of both the a and a' domains. Whereas the AGHA-PDI variant which has no catalytic activity serves as a control. The CGRC-PDI variant co-precipitated with platelet factor V in a redox-sensitive manner while there was no platelet factor V detected with the AGHA-PDI variant, thus confirming that binding of PDI to platelet-derived factor V occurs through disulfide bond exchange. Platelet factor V associates with multimerin-1 through a disulfide bond. Trapping PDI mutants also bind to multimerin-1 in a reaction requiring disulfide bond exchange. To evaluate the effect of PDI inhibition on the activation of platelet factor V, washed platelets from healthy donors were stimulated with 0.1 U/mL of thrombin in the presence of varying concentrations of isoquercetin (0 to 50 µM), which has previously been shown to inhibit PDI function. We observed a dose-dependent reduction of factor Va following platelet activation despite the fact that isoquercetin did not inhibit platelet release of PF4 or block Xa or thrombin enzymatic activity directly. We next performed a clinical study designed to determine whether oral isoquercetin inhibits thrombin generation in human subjects via its ability to inhibit platelet Va generation. Plasma samples collected from healthy participants before and 4 hours after ingestion of 1000 mg of isoquercetin (N=17). In plasma samples, post-isoquercetin platelet-dependent thrombin generation decreased by 51% compared with pre-ingestion controls (P=0.0004). Furthermore, we observed an overall 26% reduction in FVa in non-FV depleted plasma (P<0.001), which corresponded with a 53% decrease in FVa generated from platelets (P<0.001). These data confirm a significant effect of PDI inhibition on the generation of FVa following platelet activation. Considering that isoquercetin reduces platelet FVa generation and similarly inhibits platelet-dependent thrombin generation in a PDI-dependent manner, we investigated whether the addition of FVa in vitro restored platelet-dependent thrombin generation. The pre-incubation of 7 µg/mL FVa prior to stimulation with low dose thrombin restored platelet-dependent thrombin generation to within 80% baseline of pre-treatment levels. We conclude that platelet factor V is an essential substrate in mediating PDI-dependent thrombin generation on platelets and propose that PDI cleaves a disulfide bond that links platelet factor V to multimerin-1, thereby releasing platelet factor V for activation and subsequent thrombin generation. Disclosures Zwicker: Quercegen Pharma: Research Funding.


1999 ◽  
Vol 81 (04) ◽  
pp. 605-612 ◽  
Author(s):  
Dmitry V. Sakharov ◽  
Marrie Barrett-Bergshoeff ◽  
Rob T. Hekkenberg ◽  
Dingeman C. Rijken

SummaryIn a number of cases, thrombolytic therapy fails to re-open occluded blood vessels, possibly due to the occurrence of thrombi resistant to lysis. We investigated in vitro how the lysis of hardly lysable model thrombi depends on the choice of the plasminogen activator (PA) and is accelerated by ultrasonic irradiation. Lysis of compacted crosslinked human plasma clots was measured after addition of nine different PAs to the surrounding plasma and the effect of 3 MHz ultrasound on the speed of lysis was assessed.Fibrin-specific PAs showed bell-shaped dose-response curves of varying width and height. PAs with improved fibrin-specificity (staphylokinase, the TNK variant of tissue-type PA [tPA], and the PA from the saliva of the Desmodus rotundus bat) induced rapid lysis in concentration ranges (80-, 260-, and 3,500-fold ranges, respectively) much wider than that for tPA (a 35-fold range). However, in terms of speed of lysis, these three PAs exceeded tPA only slightly. Reteplase and single-chain urokinase were comparable to tPA, whereas two-chain urokinase, anistreplase, and streptokinase were inferior to tPA. In the case of fibrin-specific PAs, ultrasonic treatment accelerated lysis about 1.5-fold. For streptokinase no acceleration was observed. The effect of ultrasound correlated with the presence of plasminogen in the outer plasma, suggesting that it was mediated by facilitating the transport of plasminogen to the surface of the clot.In conclusion, PAs with improved fibrin-specificity induce rapid lysis of plasminogen-poor compacted plasma clots in much wider concentration ranges than tPA. This offers a possibility of using single-or double-bolus administration regimens for such PAs. However, it is not likely that administration of these PAs will directly cause a dramatic increase in the rate of re-opening of the occluded arteries since they are only moderately superior to tPA in terms of maximal speed of lysis. Application of high-frequency ultrasound as an adjunct to thrombolytic therapy may increase the treatment efficiency, particularly in conjunction with fibrin-specific PAs.


Sign in / Sign up

Export Citation Format

Share Document