Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884431

ABSTRACT

Ticks, lice, flees, mosquitos, leeches and vampire bats need to prevent the host's blood coagulation during their feeding process. This is primarily achieved by injecting potent anticoagulant proteins. Basophils frequently accumulate at the site of tick feeding. However, this occurs only after the second encounter with the parasite involving an adaptive immune response and IgE. To study the potential role of basophils and mast cells in the defense against ticks and other ectoparasites, we produced anticoagulant proteins from three blood-feeding animals; tick, mosquito, and leech. We tested these anticoagulant proteins for their sensitivity to inactivation by a panel of hematopoietic serine proteases. The majority of the connective tissue mast cell proteases tested, originating from humans, dogs, rats, hamsters, and opossums, efficiently cleaved these anticoagulant proteins. Interestingly, the mucosal mast cell proteases that contain closely similar cleavage specificity, had little effect on these anticoagulant proteins. Ticks have been shown to produce serpins, serine protease inhibitors, upon a blood meal that efficiently inhibit the human mast cell chymase and cathepsin G, indicating that ticks have developed a strategy to inactivate these proteases. We show here that one of these tick serpins (IRS-2) shows broad activity against the majority of the mast cell chymotryptic enzymes and the neutrophil proteases from human to opossum. However, it had no effect on the mast cell tryptases or the basophil specific protease mMCP-8. The production of anticoagulants, proteases and anti-proteases by the parasite and the host presents a fascinating example of an arms race between the blood-feeding animals and the mammalian immune system with an apparent and potent role of the connective tissue mast cell chymases in the host defense.


Subject(s)
Antithrombin Proteins/chemistry , Basophils/enzymology , Chymases/metabolism , Mast Cells/enzymology , Parasites/metabolism , Adaptive Immunity , Animals , Chemokine CCL19/chemistry , Culicidae/metabolism , Humans , Immunoglobulin E/metabolism , Leeches/metabolism , Mice , Proteolysis , Proto-Oncogene Proteins c-sis/chemistry , Ticks/metabolism
2.
Int J Mol Sci ; 22(2)2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33419227

ABSTRACT

N-linked glycosylation is a crucial post-translational modification involved in protein folding, function, and clearance. N-linked glycosylation is also used therapeutically to enhance the half-lives of many proteins. Antithrombin, a serpin with four potential N-glycosylation sites, plays a pivotal role in hemostasis, wherein its deficiency significantly increases thrombotic risk. In this study, we used the introduction of N-glycosylation sites as a tool to explore what effect this glycosylation has on the protein folding, secretion, and function of this key anticoagulant. To accomplish this task, we introduced an additional N-glycosylation sequence in each strand. Interestingly, all regions that likely fold rapidly or were surrounded by lysines were not glycosylated even though an N-glycosylation sequon was present. The new sequon in the strands of the A- and B-sheets reduced secretion, and the B-sheet was more sensitive to these changes. However, the mutations in the strands of the C-sheet allowed correct folding and secretion, which resulted in functional variants. Therefore, our study revealed crucial regions for antithrombin secretion and could potentially apply to all serpins. These results could also help us understand the functional effects of natural variants causing type-I deficiencies.


Subject(s)
Antithrombin Proteins/chemistry , Antithrombin Proteins/metabolism , Protein Conformation , Protein Processing, Post-Translational , Antithrombin III/chemistry , Antithrombin III/genetics , Antithrombin III/metabolism , Antithrombin Proteins/genetics , Circular Dichroism , Glycosylation , Humans , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Thrombosis
3.
Cell Chem Biol ; 28(1): 26-33.e8, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33096052

ABSTRACT

Despite possessing only 32 residues, the tsetse thrombin inhibitor (TTI) is among the most potent anticoagulants described, with sub-picomolar inhibitory activity against thrombin. Unexpectedly, TTI isolated from the fly is 2000-fold more active and 180 Da heavier than synthetic and recombinant variants. We predicted the presence of a tyrosine O-sulfate post-translational modification of TTI, prompting us to investigate the effect of the modification on anticoagulant activity. A combination of chemical synthesis and functional assays was used to reveal that sulfation significantly improved the inhibitory activity of TTI against thrombin. Using X-ray crystallography, we show that the N-terminal sulfated segment of TTI binds the basic exosite II of thrombin, establishing interactions similar to those of physiologic substrates, while the C-terminal segment abolishes the catalytic activity of thrombin. This non-canonical mode of inhibition, coupled with its potency and small size, makes TTI an attractive scaffold for the design of novel antithrombotics.


Subject(s)
Anticoagulants/pharmacology , Antithrombin Proteins/pharmacology , Insect Proteins/pharmacology , Thrombin/antagonists & inhibitors , Tyrosine/analogs & derivatives , Animals , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Antithrombin Proteins/chemical synthesis , Antithrombin Proteins/chemistry , Cell Line , Humans , Insect Proteins/chemical synthesis , Insect Proteins/chemistry , Molecular Structure , Thrombin/metabolism , Tsetse Flies , Tyrosine/chemical synthesis , Tyrosine/chemistry , Tyrosine/pharmacology
4.
Biotechnol Lett ; 42(1): 103-114, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31686286

ABSTRACT

OBJECTIVE: Thrombin, platelets, and plasmin are three key factors involved in hemostasis and thrombolysis. Thrombolytic therapy with clinically approved drugs is often followed by recurrent thrombosis caused by thrombin-induced platelet aggregation from the clot debris. In order to minimize these problems, new constructs were designed for the expression of recombinant staphylokinase (rSAK) and also a fusion protein composed of staphylokinase, 20 amino acids containing 2 RGD followed by tsetse thrombin Inhibitor (SAK-2RGD-TTI) in Pichia pastoris. RESULT: Modeling the tertiary structure of SAK-2RGD-TTI showed that the linker containing RGD and TTI did not interfere with proper folding of SAK. In laboratory testing, the purified SAK-2RGD-TTI (420 µg/mL) dissolved an average of 45% of the blood clot. The activity of the SAK-2RGD-TTI was also confirmed in various tests including human plasminogen activation assay, fibrin clot lysis assay, well diffusion method, activated partial thromboplastin time and platelet rich clot lysis assay. CONCLUSION: Our findings suggest that SAK-2RGD-TTI has improved therapeutic properties preventing reocclussion. It further confirms that it is practicable to assemble and produce a hybrid multifunctional protein that targets hemostatic process at various stages.


Subject(s)
Metalloendopeptidases/metabolism , Pichia/metabolism , Recombinant Fusion Proteins/metabolism , Thrombolytic Therapy/methods , Antithrombin Proteins/chemistry , Antithrombin Proteins/genetics , Antithrombin Proteins/metabolism , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Proteins/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Molecular Dynamics Simulation , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Pichia/genetics , Protein Conformation , Recombinant Fusion Proteins/genetics
5.
Biotechnol Prog ; 35(4): e2819, 2019 07.
Article in English | MEDLINE | ID: mdl-30972956

ABSTRACT

Staphylokinase (SAK) is a promising thrombolytic agent for the treatment of patients suffering from blood-clotting disorders. To increase the potency of SAK and to minimize vessel reocclusion, a new construct bearing SAK motif fused to tsetse thrombin inhibitor (TTI) via a 20-amino acid linker with 2 RGD (2 × arginine-glycine-aspartic acid inhibiting platelet aggregation via attachment to integrin receptors of platelet) was codon optimized and expressed comparatively in Pichia pastoris GS115 as a Mut+ strain and KM71H as a Muts strain. Fusion protein was optimized in terms of best expression condition and fibrinolytic activity and compared with the rSAK. Expression level of the designed construct reached up to 175 mg/L of the culture medium after 72-hr stimulation with 2.5% methanol and remained steady for 3-4 days. The highest expression was obtained at the range of 2-3% methanol. The SAK-2RGD-TT (relative activity >82%) was more active at 25-37 °C than rSAK (relative activity of 93%). Further, it showed relative activity >80% at pH ranges of 7-9. Western blot analysis showed two bands of nearly 27 and 24 kDa at ratio of 5 to 3, respectively. The specific fibrinolytic activity of the SAK-2RGD-TTI was measured as 8,269 U/mg, and 19,616 U/mg for the nonpurified and purified proteins, respectively. Deglycosylation by using tunicamycin in culture medium resulted in higher fibrinolytic activity of SAK-2RGD-TTI (2.2 fold). Consequently, compared to the rSAK, at the same equimolar proportion, addition of RGD and TTI fragments could increase fibrinolytic activity. Also, P. pastoris can be considered as an efficient host for overexpression of the soluble SAK-2RGD-TTI with high activity without requiring a complicated purification procedure.


Subject(s)
Antithrombin Proteins/pharmacology , Fibrinolytic Agents/pharmacology , Insect Proteins/pharmacology , Metalloendopeptidases/metabolism , Platelet Aggregation Inhibitors/pharmacology , Antithrombin Proteins/chemistry , Fibrinolytic Agents/chemistry , Humans , Hydrogen-Ion Concentration , Insect Proteins/chemistry , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Temperature
6.
J Biol Chem ; 292(40): 16513-16520, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28743742

ABSTRACT

Antithrombin mainly inhibits factor Xa and thrombin. The reactive center loop (RCL) is crucial for its interactions with its protease targets and is fully inserted into the A-sheet after its cleavage, causing translocation of the covalently linked protease to the opposite end of the A-sheet. Antithrombin variants with altered RCL hinge residues behave as substrates rather than inhibitors, resulting in stoichiometries of inhibition greater than one. Other antithrombin residues have been suggested to interfere with RCL insertion or the stability of the antithrombin-protease complex, but available crystal structures or mutagenesis studies have failed to identify such residues. Here, we characterized two mutations, S365L and I207T, present in individuals with type II antithrombin deficiency and identified a new antithrombin functional domain. S365L did not form stable complexes with thrombin or factor Xa, and the I207T/I207A variants inhibited both proteases with elevated stoichiometries of inhibition. Close proximity of Ile-207 and Ser-365 to the inserted RCL suggested that the preferred reaction of these mutants as protease substrates reflects an effect on the rate of the RCL insertion and protease translocation. However, both residues lie within the final docking site for the protease in the antithrombin-protease complex, supporting the idea that the enhanced substrate reactions may result from an increased dissociation of the final complexes. Our findings demonstrate that the distal end of the antithrombin A-sheet is crucial for the last steps of protease inhibition either by affecting the rate of RCL insertion or through critical interactions with proteases at the end of the A-sheet.


Subject(s)
Antithrombin Proteins/chemistry , Blood Coagulation Disorders, Inherited , Factor Xa/chemistry , Molecular Docking Simulation , Thrombin/chemistry , Amino Acid Substitution , Antithrombin Proteins/genetics , Antithrombin Proteins/metabolism , Catalytic Domain , Factor Xa/genetics , Factor Xa/metabolism , Female , Humans , Male , Mutation, Missense , Protein Domains , Protein Structure, Secondary , Thrombin/genetics , Thrombin/metabolism
7.
Bull Exp Biol Med ; 162(6): 718-721, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28429220

ABSTRACT

Coagulation and optical (based on chromogenic substrate) methods were employed to examine antithrombin activity of erythrocytes and erythrocyte-derived microvesicles isolated days 7, 14, 21, and 28 on erythrocyte storage. The erythrocyte-derived microvesicles decelerated fibrin clot formation from fibrinogen in the presence of exogenous thrombin both with and without heparin. Microvesicles reduced optical density of chromogenic substrate. These data suggest that erythrocyte-derived microvesicles display a prominent antithrombin activity, which significantly increases during erythrocyte storage.


Subject(s)
Antithrombin Proteins/chemistry , Blood Preservation/methods , Cell-Derived Microparticles/chemistry , Erythrocytes/chemistry , Fibrinogen/chemistry , Thrombin/chemistry , Adenine/chemistry , Blood Coagulation , Blood Coagulation Tests , Cells, Cultured , Chromogenic Compounds/analysis , Citrates/chemistry , Erythrocytes/cytology , Fibrin/chemistry , Glucose/chemistry , Heparin/chemistry , Humans , Phosphates/chemistry , Refrigeration/methods , Spectrophotometry
8.
Anal Chim Acta ; 947: 58-65, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27846990

ABSTRACT

Antithrombin (AT) is a plasma glycoprotein which possesses anticoagulant and anti-inflammatory properties. AT exhibits various forms, among which are native, latent and heterodimeric ones. We studied the potential of capillary electrophoresis-mass spectrometry (CE-MS) using a sheath liquid interface, electrospray ionization (ESI), and a quadrupole-time-of-flight (Q-TOF) mass spectrometer to separate and quantify the different AT forms. For CE separation, a neutral polyvinyl alcohol (PVA) coated capillary was employed. The protein conformation was preserved by using a background electrolyte (BGE) at physiological pH. A sheath liquid of isopropanol-water 50:50 (v/v) with 14 mM ammonium acetate delivered at a flow rate of 120 µL h-1 resulted in optimal signal intensities. Each AT form exhibited a specific mass spectrum, allowing unambiguous distinction. Several co-injection experiments proved that latent AT had a higher electrophoretic mobility (µep) than native AT, and that these conformers could associate to form a heterodimer during the CE analysis. The developed CE-MS method enabled the detection and quantitation of latent and heterodimeric forms in a commercial AT preparation stored at room temperature for three weeks.


Subject(s)
Antithrombin Proteins/chemistry , Electrophoresis, Capillary/methods , Mass Spectrometry/methods , Protein Multimerization , Antithrombin Proteins/isolation & purification , Models, Molecular , Protein Structure, Quaternary , Temperature
9.
Glycobiology ; 26(5): 482-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26747427

ABSTRACT

The structure of the N-linked oligosaccharides attached to antithrombin (AT) has been shown to affect its anticoagulant activity and pharmacokinetics. Human AT has biantennary complex-type oligosaccharides with the unique feature of lacking a core fucose, which affects its biological activities by changing its heparin-binding affinity. In human plasma, AT circulates as a mixture of the α-form bearing four oligosaccharides and the ß-form lacking an oligosaccharide at Asn135. However, it remains unclear how the immature high-mannose-type oligosaccharides produced by mammalian cells affect biological activities of AT. Here, we succeeded in directly comparing the activities between the high-mannose and complex types. Interestingly, although there were no substantial differences in thrombin inhibitory activity, the high-mannose type showed higher heparin-binding affinity. The anticoagulant activities were increased by heparin and correlated with the heparin-binding affinity, resulting in the strongest anticoagulant activity being displayed in the ß-form with the high-mannose type. In pharmacokinetic profiling, the high-mannose type showed a much shorter plasma half-life than the complex type. The ß-form was found to have a prolonged plasma half-life compared with the α-form for the high-mannose type; conversely, the α-form showed a longer half-life than the ß-form for the complex-type. The present study highlights that AT physiological activities are strictly controlled not only by a core fucose at the reducing end but also by the high-mannose-type structures at the nonreducing end. The ß-form with the immature high-mannose type appears to function as a more potent anticoagulant than the AT typically found in human plasma, once it emerges in the blood.


Subject(s)
Antithrombin Proteins/metabolism , Heparin/metabolism , Mannose/metabolism , Oligosaccharides/metabolism , Antithrombin Proteins/chemistry , Glycosylation , Heparin/chemistry , Humans , Mannose/chemistry , Oligosaccharides/chemistry , Protein Binding
10.
Biomed Res Int ; 2015: 630482, 2015.
Article in English | MEDLINE | ID: mdl-25866798

ABSTRACT

Physiological hemostatic balance is a coordinated outcome of counteracting coagulation and fibrinolytic systems. An imbalance of procoagulant and anticoagulant factors may result in life threatening thromboembolism. Presently, anticoagulant administration is the first line of therapy for the treatment of these conditions and several anticoagulants have been approved, including various forms of heparin. However, the polyanionic nature and multispecificity of heparin pose several complications. Generally, the polysulfated compounds with antithrombotic potential are thought to have feasible synthetic procedures with much less bleeding, thus having favourable safety profiles. Here we report the synthesis of a novel compound, trehalose octasulfate and the assessment of its anticoagulation potential. Molecular docking of trehalose and trehalose octasulfate with antithrombin showed a specificity switch in binding affinity on sulfation, where trehalose octasulfate interacts with critical residues of AT that are either directly involved in heparin binding or in the conformational rearrangement of AT on heparin binding. An in vitro analysis of trehalose octasulfate demonstrated prolonged clotting time. Lead compound when intravenously injected in occlusion induced thrombotic rats showed remarkable reduction in the size and weight of the clot at a low dose. Delay in coagulation time was observed by analysing blood plasma isolated from rats preinjected with trehalose octasulfate. A decrease in Adenosine 5'-Diphosphate (ADP) induced platelet aggregation indicated a probable dual anticoagulant and antiplatelet mechanism of action. To summarize, this study presents trehalose octasulfate as a novel, effective, dual acting antithrombotic agent.


Subject(s)
Anticoagulants , Antithrombin Proteins/chemistry , Platelet Aggregation Inhibitors , Sulfuric Acid Esters , Trehalose , Animals , Female , Male , Molecular Docking Simulation , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/pharmacology , Thrombosis/drug therapy , Trehalose/chemistry , Trehalose/pharmacology
11.
Protein Pept Lett ; 22(5): 410-8, 2015.
Article in English | MEDLINE | ID: mdl-25687119

ABSTRACT

Antithrombin inhibits blood coagulation through the interaction with serine proteases in both intrinsic and extrinsic pathways. In addition, antithrombin also shows anti-inflammatory properties, which are independent of its effects on coagulation. This work shows for the first time the cloning and sequencing of antithrombin from a snake species. This predicted protein is composed by 430 amino acids and presents about 64.5% sequence identity to human antithrombin. Biacore experiments revealed that the binding affinity of Bothrops jararaca snake antithrombin to heparin was ~30 times higher than that of human antithrombin. Furthermore, Bothrops jararaca antithrombin is more effective in preventing acute inflammation induced by carrageenan when compared to human antithrombin. Hence, the results showed herein suggest that Bothrops jararaca antithrombin can play a key role in the control of acute inflammation and that this molecule might be used as a pharmacological tool and as a prototype for drug development.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antithrombin Proteins/therapeutic use , Bothrops/genetics , Inflammation/drug therapy , Reptilian Proteins/therapeutic use , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Antithrombin Proteins/chemistry , Antithrombin Proteins/genetics , Carrageenan , Cloning, Molecular , Edema/chemically induced , Edema/drug therapy , Humans , Inflammation/chemically induced , Male , Mice , Molecular Sequence Data , Reptilian Proteins/chemistry , Reptilian Proteins/genetics , Sequence Alignment
12.
Protein Pept Lett ; 20(4): 403-11, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23016581

ABSTRACT

Endogenous angiogenesis inhibitor that specifically decreases tumor cell proliferation can be used to treat cancer since angiogenesis is required at every step of tumor progression and metastasis. Endothelial cells are the main target for the antiangiogenic therapy because they are non-transformed and easily accessible to angiogenic inhibitors. Antithrombin functions as a principal plasma protein inhibitor of blood coagulation proteinases and belongs to the family of serine protease inhibitors (serpins) which have common mechanism of inhibition. Antithrombin acquires a potent antiangiogenic activity upon conversion of the native molecule to cleaved or latent conformation. Cleaved and latent preparations of bovine and human plasma derived antithrombin inhibits capillary endothelial cell proliferation and the growth of human SK-NAS neuroblastoma and Lewis lung carcinoma tumors in mice but not the native antithrombin's. The native form of antithrombin binds with high affinity to vascular heparan sulfate proteoglycans containing a specific pentasaccharide sequence and it is this cofactor interaction that activates antithrombin to maximal rate of thrombin inhibition. Upon inhibitory complex formation with target proteinases the antithrombin undergoes stressed to relaxed transformation and lose their high affinity for pentasacchride. Low affinity relaxed conformation with reduced heparin binding like cleaved and latent are antiangiogenic but native high affinity heparin binding stressed conformation is not, indicating the critical importance of heparin affinity in antithrombin antiangiogenic function. Based on evidence of interactions of the endothelial cell growth factors bFGF (basic fibroblast growth factor) and VEGF (vascular endothelial cell growth factor) with heparin like molecule in matrix, the possibility of antiangiogenic antithrombin to interfere with endothelial cell growth and angiogenesis through heparin mediated mechanism deserves serious consideration and investigation. It is also possible that cleaved and latent conformations with reduced affinity for heparins can also induce conformational change in the antithrombin which can open an epitope on the antithrombin surface for appropriate interactions on the endothelial surface for better antiangiogenic activity. This review illustrates the potential of antithrombin and other serpin family members as endogenous antiangiogenic proteins.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antithrombin Proteins/chemistry , Antithrombin Proteins/metabolism , Antithrombins/pharmacology , Animals , Antithrombins/chemistry , Antithrombins/metabolism , Cattle , Cell Proliferation/drug effects , Endothelial Cells , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Fibroblast Growth Factor 2/metabolism , Heparin/metabolism , Humans , Protein Conformation , Serine Proteinase Inhibitors/pharmacology , Serpins/chemistry , Serpins/pharmacology , Structure-Activity Relationship
13.
J Biomol Struct Dyn ; 30(6): 684-700, 2012.
Article in English | MEDLINE | ID: mdl-22812415

ABSTRACT

The comparison between two protein structures is important for understanding a molecular function. In particular, the comparison of protein surfaces to measure their similarity provides another challenge useful for studying molecular evolution, docking, and drug design. This paper presents an algorithm, called the BetaSuperposer, which evaluates the similarity between the surfaces of two structures using the beta-shape which is a geometric structure derived from the Voronoi diagram of molecule. The algorithm performs iterations of mix-and-match between the beta-shapes of two structures for the optimal superposition from which a similarity measure is computed, where each mix-and-match step attempts to solve an NP-hard problem. The devised heuristic algorithm based on the assignment problem formulation quickly produces a good superposition and an assessment of similarity. The BetaSuperposer was fully implemented and benchmarked against popular programs, the Dali and the Click, using the SCOP models. The BetaSuperposer is freely available to the public from the Voronoi Diagram Research Center ( http://voronoi.hanyang.ac.kr ).


Subject(s)
Algorithms , Models, Molecular , Software , Amino Acid Sequence , Antithrombin Proteins/chemistry , Humans , Molecular Sequence Data , Nonlinear Dynamics , Protein Structure, Tertiary , Regression Analysis , Structural Homology, Protein , Surface Properties
14.
Mol Med ; 18: 762-70, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22481271

ABSTRACT

Mutations affecting mobile domains of antithrombin induce conformational instability resulting in protein polymerization that associates with a severe clinical phenotype, probably by an unknown gain of function. By homology with other conformational diseases, we speculated that these variants might infect wild-type (WT) monomers reducing the anticoagulant capacity. Infective polymerization of WT polymers and different P1 mutants (p.R425del, p.R425C and p.R425H) were evaluated by using native gels and radiolabeled WT monomers and functional assays. Human embryonic kidney cells expressing the Epstein-Barr nuclear antigen 1 (HEK-EBNA) cells expressing inducible (p.R425del) or two novel constitutive (p.F271S and p.M370T) conformational variants were used to evaluate intracellular and secreted antithrombin under mild stress (pH 6.5 and 39°C for 5 h). We demonstrated the conformational sensitivity of antithrombin London (p.R425del) to form polymers under mild heating. Under these conditions purified antithrombin London recruited WT monomers into growing polymers, reducing the anticoagulant activity. This process was also observed in the plasma of patients with p.R425del, p.R425C and p.R425H mutations. Under moderate stress, coexpression of WT and conformational variants in HEK-EBNA cells increased the intracellular retention of antithrombin and the formation of disulfide-linked polymers, which correlated with impaired secretion and reduction of anticoagulant activity in the medium. Therefore, mutations inducing conformational instability in antithrombin allow its polymerization with the subsequent loss of function, which under stress could sequestrate WT monomers, resulting in a new prothrombotic gain of function, particularly relevant for intracellular antithrombin. The in vitro results suggest a temporal and severe plasma antithrombin deficiency that may contribute to the development of the thrombotic event and to the clinical severity of these mutations.


Subject(s)
Antithrombin III Deficiency/metabolism , Antithrombin Proteins/chemistry , Antithrombin Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Antithrombin Proteins/genetics , Cell Line , Humans , Protein Conformation , Protein Multimerization , Protein Stability , Stress, Physiological
15.
Anim Biotechnol ; 23(2): 89-100, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22537058

ABSTRACT

Expression of recombinant pharmaceutical proteins in the mammalian mammary gland is of great interest for the medical industry. This study was designed to express recombinant human antithrombin (rhAT) in the mammary gland of rabbits by adenovirus vectors infection. Replication-defective adenovirus encoding human antithrombin complementary DNA (cDNA) was constructed and directly infused into the mammary gland of rabbits via the teat canal. The milk serum was collected from the infected mammary gland 48 h post-infection and subjected to Western blot analysis, Enzyme-linked immunosorbent assay (ELISA), and antithrombotic activity assay. In this way, the target protein was verified, and a high expression level of rhAT up to 4.8 g/L was obtained, and antithrombotic activity of the rhAT was not different than that of a standard human antithrombin protein (p > 0.05). Compared to previous attempts to produce human antithrombin in the mammary gland of transgenic animals or fractionation the plasma of blood donors, the method for rhAT expression we established would reduce production cost and further increase production efficacy.


Subject(s)
Adenoviridae/genetics , Antithrombin Proteins/biosynthesis , Mammary Glands, Animal/metabolism , Recombinant Proteins/biosynthesis , Animals , Animals, Genetically Modified , Antithrombin Proteins/analysis , Antithrombin Proteins/chemistry , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epithelial Cells , Escherichia coli , Female , Genetic Vectors/genetics , HEK293 Cells , Humans , Mammary Glands, Animal/cytology , Milk/chemistry , Rabbits , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Transfection/methods
16.
Thromb Haemost ; 107(3): 468-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22318644

ABSTRACT

The natural anticoagulant protein S contains a so-called thrombin- sensitive region (TSR), which is susceptible to proteolytic cleavage. We have previously shown that a platelet-associated protease is able to cleave protein S under physiological plasma conditions in vitro . The aim of the present study was to investigate the relation between platelet-associated protein S cleaving activity and in vivo protein S cleavage, and to evaluate the impact of in vivo protein S cleavage on its anticoagulant activity. Protein S cleavage in healthy subjects and in thrombocytopenic and thrombocythaemic patients was evaluated by immunological techniques. Concentration of cleaved and intact protein S was correlated to levels of activated protein C (APC)-dependent and APC-independent protein S anticoagulant activity. In plasma from healthy volunteers 25% of protein S is cleaved in the TSR. While in plasma there was a clear positive correlation between levels of intact protein S and both APC-dependent and APC-independent protein S anticoagulant activities, these correlations were absent for cleaved protein S. Protein S cleavage was significantly increased in patients with essential thrombocythaemia (ET) and significantly reduced in patients with chemotherapy-induced thrombocytopenia. In ET patients on cytoreductive therapy, both platelet count and protein S cleavage returned to normal values. Accordingly, platelet transfusion restored cleavage of protein S to normal values in patients with chemotherapy-induced thrombocytopenia. In conclusion, proteases from platelets seem to contribute to the presence of cleaved protein S in the circulation and may enhance the coagulation response in vivo by down regulating the anticoagulant activity of protein S.


Subject(s)
Antithrombin Proteins/metabolism , Blood Platelets/metabolism , Hematologic Neoplasms/blood , Peptide Fragments/metabolism , Protein S/metabolism , Thrombocythemia, Essential/blood , Thrombocytopenia/blood , Antithrombin Proteins/chemistry , Antithrombin Proteins/genetics , Blood Coagulation/drug effects , Blood Coagulation Tests , Blood Platelets/pathology , Catalytic Domain/genetics , Drug-Related Side Effects and Adverse Reactions , Female , Hematologic Neoplasms/complications , Hematologic Neoplasms/drug therapy , Humans , Male , Peptide Fragments/chemistry , Peptide Fragments/genetics , Platelet Transfusion , Protein C/metabolism , Protein Processing, Post-Translational/genetics , Protein S/chemistry , Protein S/genetics , Proteolysis/drug effects , Thrombocythemia, Essential/prevention & control , Thrombocytopenia/etiology , Thrombocytopenia/prevention & control
17.
Blood ; 117(23): 6347-54, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21511958

ABSTRACT

Histidine-rich protein II (HRPII) is an abundant protein released into the bloodstream by Plasmodium falciparum, the parasite that causes the most severe form of human malaria. Here, we report that HRPII binds tightly and selectively to coagulation-active glycosaminoglycans (dermatan sulfate, heparan sulfate, and heparin) and inhibits antithrombin (AT). In purified systems, recombinant HRPII neutralized the heparin-catalyzed inhibition of factor Xa and thrombin by AT in a Zn(2+)-dependent manner. The observed 50% inhibitory concentration (IC(50)) for the HRPII neutralization of AT activity is approximately 30nM for factor Xa inhibition and 90nM for thrombin inhibition. Zn(2+) was required for these reactions with a distribution coefficient (K(d)) of approximately 7µM. Substituting Zn(2+) with Cu(2+), but not with Ca(2+), Mg(2+), or Fe(2+), maintained the HRPII effect. HRPII attenuated the prolongation in plasma clotting time induced by heparin, suggesting that HRPII inhibits AT activity by preventing its stimulation by heparin. In the microvasculature, where erythrocytes infected with P falciparum are sequestered, high levels of released HRPII may bind cellular glycosaminoglycans, prevent their interaction with AT, and thereby contribute to the procoagulant state associated with P falciparum infection.


Subject(s)
Antigens, Protozoan/metabolism , Antithrombin Proteins/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Anticoagulants/pharmacology , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antithrombin Proteins/chemistry , Antithrombin Proteins/genetics , Blood Coagulation/drug effects , Blood Coagulation/genetics , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Heparin/pharmacology , Humans , Malaria, Falciparum/genetics , Metals/chemistry , Metals/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
18.
PLoS One ; 6(3): e17519, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21423730

ABSTRACT

UNLABELLED: Direct-acting fibrin(ogen)olytic agents such as plasmin have been proved to contain effective and safety thrombolytic potential. Unfortunately, plasmin is ineffective when administered by the intravenous route because it was neutralized by plasma antiplasmin. Direct-acting fibrin(ogen)olytic agents with resistance against antiplasmin will brighten the prospect of anti-thrombosis. As reported in 'Compendium of Materia Medica', the insect of Eupolyphaga sinensis Walker has been used as traditional anti-thrombosis medicine without bleeding risk for several hundreds years. Currently, we have identified a fibrin(ogen)olytic protein (Eupolytin1) containing both fibrin(ogen)olytic and plasminogen-activating (PA) activities from the beetle, E. sinensis. OBJECTIVES: To investigate the role of native and recombinant eupolytin1 in fibrin(ogen)olytic and plasminogen-activating processes. METHODS AND RESULTS: Using thrombus animal model, eupolytin1 was proved to contain strong and rapid thrombolytic ability and safety in vivo, which are better than that of urokinase. Most importantly, no bleeding complications were appeared even the intravenous dose up to 0.12 µmol/kg body weight (3 times of tested dose which could completely lyse experimental thrombi) in rabbits. It is the first report of thrombolytic agents containing both direct-acting fibrin(ogen)olytic and plasminogen-activating activities. CONCLUSIONS: The study identified novel thrombolytic agent with prospecting clinical potential because of its bi-functional merits containing both plasmin- and PA-like activities and unique pharmacological kinetics in vivo.


Subject(s)
Antithrombin Proteins/metabolism , Fibrinolysis , Plasminogen Activators/metabolism , Amino Acid Sequence , Animals , Antithrombin Proteins/administration & dosage , Antithrombin Proteins/chemistry , Antithrombin Proteins/isolation & purification , Bleeding Time , Coleoptera , Fibrinolysis/drug effects , Gastrointestinal Tract/metabolism , Hemostasis/drug effects , Humans , Hydrolysis/drug effects , Kinetics , Mice , Molecular Sequence Data , Phylogeny , Rabbits , Urokinase-Type Plasminogen Activator/pharmacology
19.
Clin Appl Thromb Hemost ; 17(3): 273-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20211924

ABSTRACT

BACKGROUND: The reactive center loop (RCL) of native antithrombin is partially inserted in the main serpin body. It must be fully exposed for optimal inhibitory function. OBJECTIVE: To test the hypothesis that P(14)-s2B interaction affects loop insertion in antithrombin. By mutating Phe(274) to Tyr(274), the objective was to introduce P(14)-s2B interaction in antithrombin. METHODS: Site-directed mutagenesis and affinity chromatography were used to obtain purified recombinant protein. Antithrombin's ability to form sodium dodecyl sulfate (SDS)-stable complex with thrombin, stoichiometry of thrombin inhibition, second-order rate constant for thrombin and factor Xa (fXa) inhibition (M(-1) s(-1)), and heparin dissociation constant (K(D); tryptophan fluorescence emission spectra) were determined. RESULTS AND CONCLUSION: A marginal, but inconclusive, difference between the wild type and the mutant was observed. The result highlights the variable effect of P(14)-s2B interaction in different serpins. Alternate hypothesis for achieving loop expulsion is proposed.


Subject(s)
Amino Acid Substitution , Antithrombin Proteins/chemistry , Catalytic Domain , Mutation, Missense , Animals , Antithrombin Proteins/genetics , Antithrombin Proteins/metabolism , Cell Line , Humans , Mutagenesis, Site-Directed , Phenylalanine/chemistry , Phenylalanine/genetics , Phenylalanine/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/metabolism
20.
J Med Chem ; 53(22): 8030-40, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-21028827

ABSTRACT

Terminal 1,6-anhydro-aminosugars (1,6-anAS) are typical structural moieties of enoxaparin, a low-molecular-weight heparin (LMWH) widely used for prevention and treatment of thrombotic disorders. In the enoxaparin manufacturing process, these modified amino sugars are formed during the ß-eliminative cleavage of heparin. To investigate the effect of terminal anAS on antithrombin (AT) binding and on inhibition of factor Xa (FXa), two octasaccharides containing modified AT-binding pentasaccharide sequences were isolated from enoxaparin. The molecular conformation of the octasaccharides terminating with N-sulfo-1,6-anhydro-D-mannosamine and N-sulfo-1,6-anhydro-D-glucosamine, respectively, has been determined both in the absence and presence of AT by NMR experiments and docking simulations. Reduced overall contacts of the terminal anAS residues with the binding region of AT induce a decrease in affinity for AT as well as lower anti-FXa activity. The anti-FXa measured either in buffer or plasma milieu does not show any significant difference, suggesting that the inhibition of anti-FXa remains specific and biologically relevant.


Subject(s)
Anticoagulants/isolation & purification , Antithrombin Proteins/chemistry , Enoxaparin/chemistry , Hexosamines/chemistry , Oligosaccharides/isolation & purification , Anticoagulants/chemistry , Anticoagulants/pharmacology , Antithrombin Proteins/metabolism , Carbohydrate Sequence , Factor Xa/chemistry , Factor Xa Inhibitors , Hexosamines/metabolism , Humans , In Vitro Techniques , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Molecular Dynamics Simulation , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Protein Binding , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...