Prevention and Therapy of Experimental Venous Thrombosis in Rabbits by Desmin 370

1998 ◽  
Vol 80 (08) ◽  
pp. 338-341 ◽  
Author(s):  
Maria Rosaria Rossiello ◽  
Miriam Barbanti ◽  
Fiorella Calanni ◽  
Nicola Semeraro ◽  
Mario Colucci

SummaryDesmin 370 (D370), a low molecular weight dermatan sulfate, has been shown to reduce the size of preformed thrombi in rats, via a mechanism largely independent of its anticoagulant activity. In the present study we investigated the therapeutic efficacy of D370 in rabbits with experimental jugular vein thrombosis. Experiments performed to evaluate the antithrombotic dosages in rabbits indicated that D370 prevented the formation of venous thrombi (Wessler model) in a dose-dependent manner with complete inhibition at 20 mg/kg. When injected to rabbits bearing a 30 min aged thrombus, D370 caused a time- and dose-dependent reduction in thrombus weight. Thrombi harvested 2 h after injection of 50 mg/kg of D370 were 71% smaller than thrombi from saline-treated rabbits and 50% smaller than pretreatment thrombi, suggesting a double effect of the drug: inhibition of thrombus accretion and reduction of the existing thrombus. Interestingly, pretreatment with the fibrinolytic inhibitor EACA (1 g/kg), significantly attenuated the therapeutic efficacy of D370, suggesting a possible involvement of the fibrinolytic system. Heparin (50 and 200 U/kg) was less active as therapeutic agent, the maximal decrease in thrombus weight, as compared to untreated rabbits, amounting to 38%. Heparin, moreover, caused a more pronounced prolongation of APTT than comparable antithrombotic dosages of D370. Our present data extend previous results on the therapeutic efficacy of D370 and underscore its potential as an alternative antithrombotic drug.

2020 ◽  
Vol 120 (04) ◽  
pp. 607-619 ◽  
Author(s):  
Lutan Zhou ◽  
Na Gao ◽  
Huifang Sun ◽  
Chuang Xiao ◽  
Lian Yang ◽  
...  

AbstractA native fucosylated glycosaminoglycan from sea cucumber Holothuria fuscopunctata (nHG), mainly branched with Fuc3S4S, exhibited potent anticoagulant activity by intrinsic tenase iXase (FIXa-FVIIIa complex) and antithrombin-dependent factor IIa (FIIa) inhibition, but also had the effects of FXII activation and platelet aggregation. For screening a selective iXase inhibitor, depolymerized nHG (dHG-1 ∼ –6) and a pure octasaccharide (oHG-8) were prepared. Like nHG, dHG-1 ∼ –6 and oHG-8 could potently inhibit iXase, and competitive binding assay indicated that dHG-5 and oHG-8 could bind to FIXa. Nevertheless, dHG-5 and oHG-8 had no effects on FXII and platelet activation. nHG, dHG-5, and oHG-8 could significantly prolong the activated partial thromboplastin time of human, rat, and rabbit plasma. In the rat deep venous thrombosis model, dHG-5 and oHG-8 showed potent antithrombotic effects in a dose-dependent manner, while the thrombus inhibition rate of nHG at high dose was markedly reduced. Additionally, dHG-5 and oHG-8 did not increase bleeding at the doses up to 10-fold of the effectively antithrombotic doses compared with nHG and low molecular weight heparin in the mice tail-cut model. Considering that dHG-5 possesses strong anti-iXase and antithrombotic activities, and its preparation process is simpler and its yield is higher compared with oHG-8, it might be a promising antithrombotic candidate.


1990 ◽  
Vol 63 (03) ◽  
pp. 505-509 ◽  
Author(s):  
Thomas Mätzsch ◽  
David Bergqvist ◽  
Ulla Hedner ◽  
Bo Nilsson ◽  
Per Østergaar

SummaryA comparison between the effect of low molecular weight heparin (LMWH) and unfragmented heparin (UH) on induction of osteoporosis was made in 60 rats treated with either UH (2 IU/ g b w), LMWH in 2 doses (2 Xal U/g or 0.4 Xal U/g) or placebo (saline) for 34 days. Studied variables were: bone mineral mass in femora; fragility of humera; zinc and calcium levels in serum and bone ash and albumin in plasma. A significant reduction in bone mineral mass was found in all heparin-treated rats. There was no difference between UH and LMWH in this respect. The effect was dose-dependent in LMWH-treated animals. The zinc contents in bone ash were decreased in all heparin-treated rats as compared with controls. No recognizable pattern was seen in alterations of zinc or calcium in serum. The fragility of the humera, tested as breaking strength did not differ between treatment groups and controls. In conclusion, if dosed according to similar factor Xa inhibitory activities, LMWH induces osteoporosis to the same extent as UH and in a dose-dependent manner. The zinc content in bone ash was decreased after heparin treatment, irrespective of type of heparin given.


1993 ◽  
Vol 128 (1) ◽  
pp. 88-94 ◽  
Author(s):  
Nobuyoshi Kokawa ◽  
Mareo Yamoto ◽  
Kenichi Furukawa ◽  
Ryosuke Nakano

We performed partial purification of low molecular weight luteinizing hormone binding inhibitor from porcine follicular fluids and examined its biological activities. Following ultrafiltration, gel filtration and anion exchange of the pooled porcine follicular fluids, low molecular weight fractions (500–10,000 MW) inhibited [125I]hLH binding to porcine granulosa cells in a dose-dependent manner. The binding inhibition kinetics study revealed that the luteinizing hormone binding inhibitor may indicate a non-competitive inhibition with [125I]hLH binding. In vitro bioassay using adult mouse testicular interstitial cells revealed that the partially purified luteinizing hormone binding inhibitor reduced ovine LH-stimulated testosterone and cAMP production in a dose-dependent manner, whereas the luteinizing hormone binding inhibitor did not affect basal production of testosterone and cAMP. The inhibitory activity was heat stable and did not disappear with activated charcoal adsorption. The results of the present study suggest that the luteinizing hormone binding inhibitor may play an important role as an ovarian non-steroidal regulator modulating the receptor binding of LH and LH-mediated steroidogenesis.


1987 ◽  
Author(s):  
B Casu ◽  
L Marchese ◽  
A Naggi ◽  
G Torri ◽  
J Fareed ◽  
...  

In order to investigate the influence of charge distribution and chain length on the biological properties of sulfated polysaccharides, additional sulfate groups were introduced into the galactosaminoglycans, chondriotin sulfate and dermatan sulfate. Using a flexible method (with sulfuric acid and chlorosulfonic acid) for concurrent sulfation and controlled depolymerization, numerous products were obtained and characterized by chemical, enzymatic and nuclear magnetic resonance spectroscopic methods. The biologic actions of these products were profiled in both in vitro and in vivo assays for antithrombotic activity. Despite a weaker in vitro anticoagulant activity, low molecular weight over sulfated galactosaminoglycans produced significant dose-dependent antithrombotic actions in animal models which were similar to the actions observed with oversulfated low molecular weight heparins. These results suggest that a significant antithrombotic activity can be elicited through non-specific interactions of polysulfates with cellular and plasma components, and that clusters of sulfate groups such as the 4-6 disulfate group on D-galactosaminoglycan residues may be important for these interactions. Furthermore, these results, also suggest that supersulfation of glycosaminogly-cans results in products with biologic activity distinct from the native material.


2020 ◽  
Vol 981 ◽  
pp. 258-264
Author(s):  
Nurul Haida Idrus ◽  
Nina Suhaity Azmi ◽  
Che Nur Mazadillina Che Zahari ◽  
Solachuddin Jauhari Arief Ichwan

Glycosaminoglycans (GAGs) are long unbranched polysaccharide that composed of repeating disaccharide units. They are classified into heparan sulfate (HS), heparin, chondroitin sulfate (CS), dermatan sulfate (DS), keratan sulfate (KS) and hyaluronic acid (HA). During the last decade, demand of GAGs were getting increased due to their potential uses. Vertebrate animal, commonly cartilaginous mammalian tissue, were potential producer of GAGs and have the higher number of biological activities extracted from sea bass waste. Sea bass waste from Lates calcarifer was used as the raw material to extract crude GAGs. Different part of sea bass waste such as, gills, viscera and air bladders were used. The higher content of crude GAGs in sea bass waste was used in cytotoxic and inflammatory study. Different concentration of extract GAGs from gills were used ranging between 0.16-20 mg/mL. GAGs from sea bass waste (gills) showed dose-dependent cytotoxic activity towards MCF-7 cell line in lower concentration. Meanwhile, for anti-inflammatory study GAGs from sea bass waste (gills) showed dose-dependent manner and also reduce NO production in LPS-stimulated cells. This research study concluded that, GAGs from sea bass waste are the alternative source that can be used for cancer and inflammation study.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 22-22 ◽  
Author(s):  
Joshuaine Toth ◽  
Guanfa Gan ◽  
Joanne van Ryn ◽  
Holly Dursema ◽  
Jennifer Isler ◽  
...  

Abstract Abstract 22 Background: The objective of this study is to determine the pharmacokinetics (PK) and pharmacodynamics (PD) of dabigatran (a small molecule thrombin inhibitor) and its antidote (a humanized Fab against dabigatran) in the monkey and to develop a combined mechanistic mathematical model to describe the data. Methods: There were three groups: control, antidote alone and dabigatran etexilate (DE) + antidote. Rhesus monkeys (n = 2/group) received either 12 mg/kg/day of DE or vehicle orally on Days 1–4, 15–18 and 29–32 with a single IV dose of the antidote administered 90 minutes after DE on Days 4, 18 and 32. Doses of the antidote were 30, 90 or 175 mg/kg, respectively. PK parameters of the antidote and sum dabigatran (dabigatran plus its glucuronides) were determined after measurements of plasma concentrations. Coagulation activity was measured using a diluted thrombin time assay to determine the activity of the unbound sum dabigatran. Results: The PK of the antidote were not affected by dabigatran. Clearance of the antidote was low (0.87 mL/min/kg) and steady-state volume of distribution was small (0.06 L/kg), indicating that the antidote was mostly restricted to plasma. The plasma profile of the antidote was bi-phasic with a short initial phase t1/2 of 0.4 hour (h) and a terminal phase t1/2 of 4.3 h. Immediately after antidote dosing, plasma concentrations of sum dabigatran increased, a consequence of the rapid redistribution of dabigatran and its glucuronides from tissue to plasma due to binding to the antidote. Complete reversal of dabigatran's anticoagulant activity was observed immediately after antidote dosing at all three dose levels, as measured by the diluted thrombin time assay, which indicates that all dabigatran was bound to the antidote. The degree to which this reversal effect was maintained over an extended period (24 h) was dose-dependent. A mechanistic ordinary differential equation model, based on the mass action kinetics for describing the distribution, binding and elimination of dabigatran and its antidote, was developed by combining the PK models for dabigatran and the antidote and adding the binding interaction (1:1 stoichiometry) between the two compounds. The distribution and elimination parameters of the dabigatran-antidote complex were assumed to be the same as those of the antidote, based on similar measured PK parameters of the antidote with and without dabigatran in the monkey. The combined PK/PD model of dabigatran and antidote was able to describe the in vivo PK/PD data observed in monkeys. Conclusion: The dabigatran-specific antidote successfully reversed the anticoagulant activity of dabigatran in the monkey in a dose-dependent manner, and our combined mathematical model accurately describes monkey PK/PD data of sum dabigatran and its antidote. Insights gained from this model will be used to guide model development for clinical trials. Disclosures: Toth: Boehringer Ingelheim: Employment. Gan:Boehringer Ingelheim: Employment. van Ryn:Boehringer Ingelheim: Employment. Dursema:Boehringer Ingelheim: Employment. Isler:Boehringer Ingelheim: Employment. Coble:Boehringer Ingelheim: Employment. Burke:Boehringer Ingelheim: Employment. Lalovic:Boehringer Ingelheim: Employment. Olson:Boehringer Ingelheim: Employment.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2314-2314
Author(s):  
Jin Im ◽  
Amanda Cernosek ◽  
Anna Sergeeva ◽  
Jeffrey J. Molldrem ◽  
Lu Sijie

Abstract PR1 is a HLA-A2 restricted immune-dominant peptide derived from proteinase 3 and neutrophil elastase - leukemia associated self-antigens aberrantly over-expressed in myeloid leukemia. The persistent PR1/HLA-A2 specific cytotoxic T cell responses are associated with prolonged remission from myeloid leukemia, suggesting that PR1/HLA-A2 would be a potent target for T cell based immunotherapy. Previously, we have successfully developed a high affinity T cell receptor (TCR)-like antibody specific for the PR1/HLA-A2 complex (h8F4), and demonstrated that h8F4 mediated specific lyses of myeloid leukemia cells invitro via antibody dependent cell mediated cytotoxicity (ADCC), and significantly reduced leukemic blasts invivo in murine xenograft model of treatment refractory acute myeloid leukemia. We hypothesize that the therapeutic efficacy of h8F4 would be enhanced by combining T cell mediated cytotoxicity through a novel antibody engineering technology, called, Bi-Specific T cell Engager (BiTE) specific for T cells and tumor associated antigens. The BiTE molecules redirect effector function of polyclonal T cells against the tumor leading to lyses of tumor cells, and early clinical data show promising therapeutic efficacy of this approach. In the current study, we constructed Bi-Specific T cell Engaging antibody: h8F4-BiTE, a single chain Fragment of variable domain (scFv) of h8F4 conjugated with scFv of anti-CD3 antibody – OKT3, and produced h8F4-BiTE proteins using eukaryotic expression system. The h8F4-BiTE proteins retained the similar binding specificity to PR1/HLA-A2 as compared to the parent antibody (h8F4), demonstrated by flowcytometry based binding assays with a series of alanine substituted PR1 peptide/HLA-A2 complexes on T2 cells. The 8F4-BiTE bound to native and endogenous PR1/HLA-A2 complexes presented by HLA-A2 positive or transduced myeloid cell lines such as K562, THP-1, and HL-60. In addition, h8F4-BiTE showed comparable binding affinity to CD3 moiety of T cells. Lastly, h8F4-BiTE proteins activated T cells in a PR1/HLA-A2 specific and dose dependent manner, shown by increased expression of activation markers such as CD69 when co-cultured with PR1-pulsed T2 target cells in various concentrations of h8F4-BiTE proteins. Importantly, the PR1/HLA-A2 specific activation of T cells by h8F4-BiTE proteins was equally efficient among all subsets of T cells regardless of their phenotypes, e.g., CD4 vs CD8, and naïve vs memory, suggesting that h8F4-BiTE would be a highly potent inducer of cytotoxic T cells against PR1/HLA-A2 positive myeloid leukemia. Moreover, we generated multiple h8F4-BiTE variants with specific mutations in frameworks and/or complement determining regions (CDRs), and compared binding affinity and function to wildtype h8F4-BiTE. The h8F4-BiTE variants indeed improved specificity to PR1/HLA-A2 complex by complete elimination of residual non-specific binding to HLA-A2, and increased the binding affinity to CD3 by 10-folds resulting in dramatic 50 fold increase in potency to activate T cells. In conclusion, we have developed a novel immunetherapeutics: h8F4-BiTE targeting PR1/HLA-A2 myeloid leukemia antigen. We demonstrated high binding affinity and specificity of h8F4-BiTE to both PR1/HLA-A2 on myeloid leukemia cells and CD3 on T cells and in vitro activation of T cells in the presence of PR1/HLA-A2 expressing target cells in a dose dependent manner. Finally, we improved antigen specificity and functional activity of h8F4-BiTE via selective site-directed mutagenesis. Further preclinical evaluation of therapeutic efficacy is thus warranted using animal in vivo animal treatment model with NOD/SCID/IL2Rγ −/− mice xenografted with human myeloid leukemia cells. This unique TCR-like therapeutic agent adds an additional mechanism of action to PR1/HLA-A2 specific antibody (h8F4) and promises to enhance T cell function against myeloid leukemia. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1992 ◽  
Vol 79 (2) ◽  
pp. 430-438
Author(s):  
TC Wun

Lipoprotein-associated coagulation inhibitor (LACI) is a plasma-derived protein that inhibits tissue factor (TF)/factor VIIa-induced coagulation in a factor Xa-dependent manner. The roles of endogenous plasma LACI and exogenously added LACI and heparin, in the regulation of coagulation, initiated via the intrinsic and extrinsic pathways, were studied using the activated partial thromboplastin time (APTT) and the modified prothrombin time (PT) assays, respectively. Both LACI- depleted plasma and normal plasma have identical APTTs and similar prolongations of the APTT in response to heparin; both are fully anticoagulated (arbitrarily defined as clotting times of greater than 1 hour) at similar concentrations of heparin. These results indicate that heparin is an effective anticoagulant when coagulation is initiated by the intrinsic pathway and that endogenous LACI is not significantly involved in the regulation of this pathway. The PT of normal plasma is only marginally longer than that of LACI-depleted plasma in the absence of heparin, suggesting that endogenous plasma LACI has a very limited capacity to inhibit TF-induced clotting. However, in the presence of heparin, the PTs of LACI-depleted plasma and normal plasma are different. Prolongation of the PT occurred only moderately and linearly with increasing concentrations of heparin in LACI-depleted plasma. In contrast, normal plasma showed a greater extent of PT prolongation in response to heparin and the plasma became fully anticoagulated at a certain threshold concentration of heparin. These results suggest that LACI serves as a cofactor for heparin and thus greatly enhances the inhibition of TF-induced coagulation. LACI-depleted plasma was supplemented with purified recombinant LACI and/or heparin and the effects on TF-induced clotting were studied. A combination of LACI and heparin greatly enhanced anticoagulation compared with LACI or heparin alone. Many sulfated polysaccharides were also found to enhance the LACI-dependent inhibition of TF-induced clotting. By weight, the relative potencies of these compounds are: low molecular weight heparin (mean Mr, 5,100) greater than unfractionated heparin greater than low molecular weight heparin (mean Mr, 3,700) greater than pentosan polysulfate greater than dermatan sulfate greater than dextran sulfate greater than heparan sulfate. Based on the above results, it is concluded that LACI is a cofactor for heparin in the inhibition of TF- induced clotting and that LACI and sulfated polysaccharides act synergistically in whole plasma.


1993 ◽  
Vol 70 (03) ◽  
pp. 443-447 ◽  
Author(s):  
Benilde Cosmi ◽  
Giancarlo Agnelli ◽  
Edward Young ◽  
Jack Hirsh ◽  
Jeffrey Weitz

SummaryThe aim of this study was to investigate the mechanism by which the anticoagulant activity of dermatan sulfate (DS) is increased by low molecular weight heparin (LMWH). In platelet poor plasma, LMWH enhances the effect of DS on thrombin (IIa) inhibition as determined by thrombin clotting times and with a chromogenic substrate assay. Analysis of the results of the chromogenic assays using either the algebraic fractional or the graphic isobole method suggests that LMWH has an additive effect on the anti-IIa activity of DS. This additive effect was lost when the experiments were repeated in plasma immunodepleted of antithrombin III (ATIII), indicating that the anti-IIa activity of LMWH is ATIII-dependent. To further explore the mechanism of the interaction between LMWH and DS, 125I-labeled IIa was added to plasma in the presence or absence of DS and/or LMWH and the formation of IIa-inhibitor complexes was assessed using SDS-PAGE followed by autoradiography. DS addition selectively increases the formation of heparin cofactor II (HCII)-IIa complexes, whereas LMWH enhances ATIII-IIa complex generation. Compared to plasma containing DS alone, the formation of ATIII-IIa complexes also is increased when the combination of DS and LMWH is added. These findings suggest that the additive effect of LMWH on the anti-IIa activity of DS reflects their different modes of IIa inhibition; DS potentiates IIa inhibition by HCII, while LMWH catalyses ATIII-dependent IIa inactivation. The potential clinical significance of these findings requires further investigation.


1986 ◽  
Vol 55 (03) ◽  
pp. 309-313 ◽  
Author(s):  
J-M Freyssinet ◽  
Marie-Louise Wiesel ◽  
Josiane Gauchy ◽  
B Boneu ◽  
J-P Cazenave

SummaryAn anticoagulant activity was isolated from the plasma of a patient with a strong lupus-like anticoagulant using gel filtration by high performance liquid chromatography. IgM were detected in this anticoagulant fraction which exhibited specificity towards 50% phosphatidylcholine - 50% phosphatidylserine vesicles and cardiolipin. These phospholipids were able to produce an apparent 3-fold enhancement of purified human protein C activation by human a-thrombin in the presence of purified human placenta thrombomodulin. In the absence of phospholipid, the anticoagulant fraction had no effect on thrombomodulin activity. The anticoagulant fraction could neutralize the enhancement of thrombomodulin activity by phospholipid in a dose-dependent manner. This study suggests that the neutralization of phospholipid might result in a reduced activation of protein C which could be responsible for the occurrence of thrombotic complications in a proportion of patients with lupus anticoagulants.


Sign in / Sign up

Export Citation Format

Share Document