Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Thromb Res ; 230: 18-26, 2023 10.
Article in English | MEDLINE | ID: mdl-37607435

ABSTRACT

INTRODUCTION: Hereditary antithrombin (AT) deficiency type I causes venous thrombosis due to decreased levels of AT antigen in the blood. We identified one novel and one known abnormal variant in two unrelated Japanese families with venous thrombosis. In this study, we analyzed the mechanism by which these abnormal variants cause type I AT deficiency. MATERIALS AND METHODS: Wild-type and variant AT expression vectors were constructed and transiently expressed in human embryonic kidney 293 cells, and AT antigen levels and N-glycosylation of cell lysates and culture medium were evaluated by western blot analysis. Subcellular co-localization of AT was also examined using confocal microscopy, and chase experiments with cycloheximide and MG132 were performed to investigate the degradation pathway of AT variants. RESULTS: Genetic analysis identified a novel variant, c.613delC (p.Leu205Trpfs⁎79), and the known variant c.283T>C (p.Tyr95His). These AT variants exhibited significantly reduced extracellular secretion compared with the wild-type; N-glycosylation of the AT protein was normal. Co-localization analysis suggested that the transport of these abnormal AT proteins to the Golgi apparatus was impaired. The c.613delC variant was degraded early by the proteasome, suggesting that the c.283T>C variant is stored in the endoplasmic reticulum (ER). CONCLUSIONS: The AT variants identified here synthesize abnormal AT proteins that exhibit suppressed secretion and impaired transport from the ER to the Golgi apparatus. These results provide clues that could help elucidate the mechanism of type I AT deficiency and facilitate therapy development.


Subject(s)
Antithrombin III Deficiency , Venous Thrombosis , Humans , Antithrombins , Antithrombin Proteins , Antithrombin III/genetics , Antithrombin III Deficiency/genetics , Venous Thrombosis/genetics
2.
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
3.
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
4.
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
5.
Clin Appl Thromb Hemost ; 26: 1076029620981630, 2020.
Article in English | MEDLINE | ID: mdl-33332196

ABSTRACT

In Japan, the dose of the new recombinant antithrombin III concentrate (rAT-gamma) is titrated according to patient body weight (BW), while conventional plasma-derived antithrombin concentrates (AT) are administered as a fixed dose. Therefore, it is anticipated that rAT-gamma could produce better treatment effects than AT. The aim of this study was to compare the organ protective effects of doses of rAT-gamma and AT administered in clinical practice for septic disseminated intravascular coagulation (DIC) and multiple organ failure. This study was performed at a single university hospital in Japan. A total of 49 patients with antithrombin deficiency secondary to septic DIC who were administered either rAT-gamma (n = 26) or AT (n = 23) were retrospectively analyzed to assess the dose of supplemental antithrombin concentrates, plasma antithrombin activity, Japanese Association for Acute Medicine (JAAM)-DIC score, and modified Sequential Organ Failure Assessment (SOFA) score on days 0, 3 and 6. The AT-equivalent dose per kg BW of rAT-gamma (equal to the initial rAT-gamma dose per kg BW divided by 1.2) was significantly higher than the dose per kg BW of AT (AT 23.4 ± 5.1 vs. rAT 28.9 ± 3.9 IU/kg/day; P < 0.001). Consequently, serial increases in plasma antithrombin levels occurred more rapidly in the rAT-gamma group (P = 0.036). JAAM DIC and modified SOFA scores revealed significantly greater improvement in the rAT versus the AT group (JAAM DIC score: P = 0.042, mSOFA score: P = 0.005). The results of this study suggest that AT supplementation adjusted for patient BW might further improve septic DIC and multiple organ failure.


Subject(s)
Antithrombin Proteins/therapeutic use , Antithrombins/therapeutic use , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Sepsis/complications , Aged , Female , Humans , Male , Middle Aged , Recombinant Proteins/therapeutic use , Retrospective Studies
6.
Am J Obstet Gynecol ; 223(5): 739.e1-739.e13, 2020 11.
Article in English | MEDLINE | ID: mdl-32780999

ABSTRACT

BACKGROUND: Despite expectant management, preeclampsia remote from term usually results in preterm delivery. Antithrombin, which displays antiinflammatory and anticoagulant properties, may have a therapeutic role in treating preterm preeclampsia, a disorder characterized by endothelial dysfunction, inflammation, and activation of the coagulation system. OBJECTIVE: This randomized, placebo-controlled clinical trial aimed to evaluate whether intravenous recombinant human antithrombin could prolong gestation and therefore improve maternal and fetal outcomes. STUDY DESIGN: We performed a double-blind, placebo-controlled trial at 23 hospitals. Women were eligible if they had a singleton pregnancy, early-onset or superimposed preeclampsia at 23 0/7 to 30 0/7 weeks' gestation, and planned expectant management. In addition to standard therapy, patients were randomized to receive either recombinant human antithrombin 250 mg loading dose followed by a continuous infusion of 2000 mg per 24 hours or an identical saline infusion until delivery. The primary outcome was days gained from randomization until delivery. The secondary outcome was composite neonatal morbidity score. A total of 120 women were randomized. RESULTS: There was no difference in median gestational age at enrollment (27.3 weeks' gestation for the recombinant human antithrombin group [range, 23.1-30.0] and 27.6 weeks' gestation for the placebo group [range, 23.0-30.0]; P=.67). There were no differences in median increase in days gained (5.0 in the recombinant human antithrombin group [range, 0-75] and 6.0 for the placebo group [range, 0-85]; P=.95). There were no differences between groups in composite neonatal morbidity scores or in maternal complications. No safety issues related to recombinant human antithrombin were noted in this study, despite the achievement of supraphysiological antithrombin concentrations. CONCLUSION: The administration of recombinant human antithrombin in preterm preeclampsia neither prolonged pregnancy nor improved neonatal or maternal outcomes.


Subject(s)
Antithrombin Proteins/therapeutic use , Cesarean Section/statistics & numerical data , Gestational Age , Pre-Eclampsia/drug therapy , Administration, Intravenous , Adolescent , Adult , Delivery, Obstetric/statistics & numerical data , Double-Blind Method , Female , Fetal Distress/epidemiology , Humans , Infant, Premature, Diseases/epidemiology , Infant, Small for Gestational Age , Middle Aged , Neonatal Sepsis/epidemiology , Perinatal Mortality , Pre-Eclampsia/blood , Pre-Eclampsia/physiopathology , Pregnancy , Pregnancy Trimester, Second , Pregnancy Trimester, Third , Prospective Studies , Recombinant Proteins , Young Adult
7.
J Thromb Thrombolysis ; 50(3): 580-586, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32648093

ABSTRACT

The outbreak of novel coronavirus disease 2019 (COVID-19) has now become a global pandemic. Coagulopathy has been reported widely in critically ill COVID-19 patients and was related to high mortality. However, the comprehensive coagulation profiles have not been examined and the underlying mechanism of the coagulopathy in COVID-19 patients is unclear. To study the coagulation profiles of routine hemostasis tests, natural anticoagulants, coagulant factors and antiphospholipid antibodies in critically ill COVID-19 patients. This single-center and cross-section study included 19 patients with COVID-19, who were admitted to intensive care unit (ICU) at Tongji hospital in Wuhan, China, from Feb 23 to Mar 3, 2020. Demographic data, laboratory parameters, treatments and clinical outcomes of the patients were collected and analyzed. The final date of follow-up was Mar 31, 2020. In this study, 12 thrombotic events occurred in 9 patients, including 4 cerebral infarctions, 7 acro-ischemia and 1 internal jugular vein thrombosis. The common abnormalities of routine coagulation tests included evelated D-Dimer level (100%), prolonged prothrombin time (73.7%) and hyperfibrinogenemia (73.7%). The median activities of natural anticoagulants including protein C, protein S and antithrombin were all below the normal range. Factor VIII activities were significantly above normal range (median value 307%, IQR 198-441) in all patients. Factor V and factor VII activities were significantly lower in near-terminal stage patients. Anti-phospholipid antibodies were present in 10 patients. Strikingly, 4 cerebral infarction events were in patients had anti-phospholipid antibodies of multiple isotypes. Sustained hypercoagulable status and thrombotic events were common in critically ill patients with COVID-19. The low activities of natural anticoagulants, elevated factor VIII level and the presence of antiphospholipid antibodies, together, may contribute to the etiopathology of coagulopathy in COVID-19 patients.


Subject(s)
Antibodies, Antiphospholipid/blood , Betacoronavirus/pathogenicity , Blood Coagulation Factor Inhibitors/blood , Blood Coagulation Factors/analysis , Blood Coagulation , Coronavirus Infections/blood , Pneumonia, Viral/blood , Thrombosis/blood , Aged , Antithrombin Proteins/analysis , Biomarkers/blood , COVID-19 , China , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Critical Illness , Cross-Sectional Studies , Female , Fibrin Fibrinogen Degradation Products/analysis , Host-Pathogen Interactions , Humans , Male , Middle Aged , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , Protein C/analysis , Protein S/analysis , Risk Factors , SARS-CoV-2 , Thrombosis/diagnosis , Thrombosis/virology
8.
Arterioscler Thromb Vasc Biol ; 40(7): 1748-1762, 2020 07.
Article in English | MEDLINE | ID: mdl-32404004

ABSTRACT

OBJECTIVE: Native and latent conformers of AT (antithrombin) induce anti-inflammatory and proapoptotic signaling activities, respectively, in vascular endothelial cells by unknown mechanisms. Synd-4 (syndecan-4) has been identified as a receptor that is involved in transmitting signaling activities of AT in endothelial cells. Approach and Results: In this study, we used flow cytometry, signaling assays, immunoblotting and confocal immunofluorescence microscopy to investigate the mechanism of the paradoxical signaling activities of high-affinity heparin (native) and low-affinity heparin (latent) conformers of AT in endothelial cells. We discovered that native AT binds to glycosaminoglycans on vascular endothelial cells via its heparin-binding D-helix to induce anti-inflammatory signaling responses by recruiting PKC (protein kinase C)-δ to the plasma membrane and promoting phosphorylation of the Synd-4 cytoplasmic domain at Ser179. By contrast, the binding of latent AT to endothelial cells to a site(s), which is not competed by the native AT, induces a proapoptotic effect by localizing PKC-δ to the perinuclear/nuclear compartment in endothelial cells. Overexpression of a dominant-negative form of PKC-δ resulted in inhibition of anti-inflammatory and proapoptotic signaling activities of both native and latent AT. CONCLUSIONS: These results indicate that the native and latent conformers of AT may exert their distinct intracellular signaling effects through differentially modulating the subcellular localization of PKC-δ in endothelial cells.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antithrombin Proteins/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Protein Kinase C-delta/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Active Transport, Cell Nucleus , Cell Line , Endothelial Cells/enzymology , Endothelial Cells/pathology , Heparan Sulfate Proteoglycans/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase C-delta/genetics , Signal Transduction , Syndecan-4/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
9.
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
10.
Pediatr Blood Cancer ; 66(7): e27729, 2019 07.
Article in English | MEDLINE | ID: mdl-30938058

ABSTRACT

BACKGROUND: L-asparaginase is a cornerstone treatment for children with acute lymphoblastic leukemia (ALL). However, immune reaction to the drug may increase the clearance or impair the function of L-asparaginase and reduces its therapeutic efficacy. The objective of this study was to identify potential plasma proteins that could be used as proxies for L-asparaginase activity. METHODS: Fibrinogen, von Willebrand factor antigen (VWF:Ag), total protein, and albumin levels as well as antithrombin (AT) and L-asparaginase activities were measured in 97 children with ALL treated for prolonged period of time with L-asparaginase. Binary logistic regression and a receiver operating characteristic (ROC) curve analysis were performed to evaluate the predictive value of plasma proteins for L-asparaginase activity. RESULTS: Median E. coli L-asparaginase activity was 220 IU/L (range, 0-1308) throughout the treatment period. L-asparaginase activity was below 100 IU/L in 23% of measured samples. L-asparaginase activity was inversely associated with AT activity, fibrinogen, total protein, and albumin levels (r = -0.63, -0.62, -0.57, and -0.45, respectively; P < 0.0001), but not with VWF:Ag. ROC curve analyses showed an intermediate accuracy of AT activity (area under the ROC curve [AUC] = 0.77) to detect specimens with subtherapeutic level of L-asparaginase. An optimal accuracy was found when AT and fibrinogen were combined (AUC = 0.82; sensitivity = 75%; specificity = 82%; positive predictive value = 55%; negative predictive value = 92%) with cutoff values of 0.73 IU/mL and 1.85 g/L, respectively. CONCLUSIONS: AT combined with fibrinogen levels could be used as a proxy to identify patients with therapeutic level of L-asparaginase activity in the absence of real-time asparaginase measurement during prolonged exposure to L-asparaginase.


Subject(s)
Antithrombin Proteins/metabolism , Asparaginase/administration & dosage , Fibrinogen/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Time Factors
11.
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
12.
Thromb Res ; 171: 1-6, 2018 11.
Article in English | MEDLINE | ID: mdl-30216821

ABSTRACT

INTRODUCTION: Injury and loss of the endothelial glycocalyx occur during the early phase of sepsis. We previously showed that antithrombin has a protective effect on this structure in vitro. Here, we investigated the possible protective effects of antithrombin in an animal model of sepsis. METHODS: Wistar rats were injected with endotoxin, and circulating levels of syndecan-1, hyaluronan, albumin, lactate and other biomarkers were measured in an antithrombin-treated group and an untreated control group (n = 6 in each group). Intravital microscopy was used to observe leukocyte adhesion, microcirculation, and syndecan-1 staining. RESULTS: The circulating levels of syndecan-1 and hyaluronan were significantly reduced in the antithrombin-treated group, compared with the untreated controls. Lactate levels and albumin reduction were significantly attenuated in the antithrombin-treated group. Intravital microscopic observation revealed that both leukocyte adhesion and blood flow were better maintained in the treatment group. The syndecan-1 lining was disrupted after endotoxin treatment, and this derangement was attenuated by treatment with antithrombin. CONCLUSION: Antithrombin effectively maintained microcirculation and vascular integrity by protecting the glycocalyx in a rat sepsis model.


Subject(s)
Antithrombin Proteins/therapeutic use , Antithrombins/therapeutic use , Endothelium, Vascular/drug effects , Glycocalyx/drug effects , Sepsis/drug therapy , Sepsis/pathology , Animals , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Endotoxins/immunology , Glycocalyx/immunology , Glycocalyx/pathology , Hyaluronic Acid/blood , Hyaluronic Acid/immunology , Inflammation/blood , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Microcirculation/drug effects , Rats, Wistar , Sepsis/blood , Sepsis/immunology , Syndecan-1/blood , Syndecan-1/immunology
13.
J Clin Pathol ; 71(7): 614-619, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29331940

ABSTRACT

AIMS: Dysfunctional prothrombin residue Arg596 associated mutation has been found to precipitate venous thromboembolism (VTE). In the current study we investigated the prevalence of Arg596 associated mutations in Chinese patients with VTE and explored the functional impact of Arg596Gln mutation on coagulation function in affected patients. METHODS: Prothrombin clotting activity was measured in 267 unrelated patients with unprovoked VTE. Patients with moderately decreased activities underwent further analysis of the F2 gene. Prothrombin amidolytic activity and antigen levels were detected in mutation carriers. Specific family members were investigated about their VTE histories and clinical phenotypes. The thrombin generation test (TGT) was used to evaluate thrombin function and antithrombin resistance assay was applied to assess the extent of impaired antithrombin inhibition of mutation carriers. RESULTS: Two heterozygous mutation carriers of prothrombin Arg596Gln were identified, both of whom had moderately decreased clotting activities but normal amidolytic activities and antigen levels. Among the families of the two probands, nine out of 13 mutation carriers experienced episodes of VTE. TGTs showed that patients had elevated endogenous thrombin potential and prolonged start tail time. Thrombin generation could be inhibited in the presence of thrombomodulin. The thrombin Arg596Gln variant in patients' plasma presented strong resistance to antithrombin inhibition. CONCLUSION: Prothrombin Arg596Gln mutation is a risk factor for Chinese patients with VTE due to its moderately decreased clotting activity but strong resistance to antithrombin inhibition. Prothrombin clotting activity screening and its encoding gene sequencing should be considered in patients with VTE when other established risk factors are absent.


Subject(s)
Blood Coagulation/genetics , DNA Mutational Analysis , Genetic Testing/methods , Mutation , Prothrombin/genetics , Venous Thromboembolism/genetics , Adult , Antithrombin Proteins/metabolism , Asian People/genetics , Blood Coagulation Tests , Case-Control Studies , China/epidemiology , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Heredity , Heterozygote , Humans , Male , Middle Aged , Pedigree , Phenotype , Risk Factors , Venous Thromboembolism/blood , Venous Thromboembolism/diagnosis , Venous Thromboembolism/ethnology
14.
Int J Lab Hematol ; 40(3): 251-257, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29356352

ABSTRACT

INTRODUCTION: Factor VII activation occurs postprandially. A proportion of activated factor VII (VIIa) circulates in complex with antithrombin (VIIaAT). Our primary objective was to assess the effects of postprandial lipemia on circulating VIIaAT, procoagulant phospholipid (PPL) activity, and thrombin generation. METHODS: Plasma samples from postmyocardial infarction patients (n = 40) and controls (n = 39) were taken before and at 3 and 6 hours during a standardized oral fat tolerance test (OFTT). Fasting PPL activity measurements were also made in a second cohort of 108 postinfarction patients and 109 controls. VIIaAT was analyzed with the Asserachrom VIIaAT ELISA, PPL activity with the STA-Procoag-PPL kit, and thrombin generation with calibrated automated thrombogram with PRP-Reagent as trigger (all Diagnostica Stago products). RESULTS: Postprandially, VIIaAT increased in all samples without significant case-control differences in the overall response during the OFTT. Thrombin generation measures peak height and velocity, and PPL activity, were marginally affected by the test meal in the controls. Levels of all patient baseline measures were significantly different from controls, indicating a more hypercoagulable state, and these differences were maintained throughout the OFTT. Fasting samples from cases showed higher PPL activity than control samples. CONCLUSION: Viewing VIIaAT quantitation as a surrogate for TF activity measurement, postprandial increase in VIIaAT may reflect a mechanism that adds to the cardiovascular risk associated with postprandial lipemia. On the other hand, the impact of postprandial lipemia on PPL activity and thrombin generation seems to be minor.


Subject(s)
Antithrombin III/metabolism , Factor VIIa/metabolism , Hyperlipidemias/metabolism , Phospholipids/metabolism , Postprandial Period , Thrombin/biosynthesis , Adult , Antithrombin Proteins/metabolism , Cardiovascular Diseases/etiology , Case-Control Studies , Female , Humans , Hyperlipidemias/complications , Male , Middle Aged , Myocardial Infarction , Protein Binding
15.
Int J Hematol ; 107(4): 490-494, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29071478

ABSTRACT

Inherited antithrombin (AT) deficiency is one of the most clinically significant forms of congenital thrombophilia and follows an autosomal dominant mode of inheritance. We analyzed SERPINC1 in a patient who developed deep-vein thrombosis and low AT activity during pregnancy, and identified a novel missense mutation c.259A>G (p.Asn87Asp; N87D). Surprisingly, analysis of the parents' DNA showed that they did not possess this mutant, and thus, it may have been due to a de novo mutation. We also expressed this mutant AT protein in COS-1 cells and compared its intracellular localization and intracellular and extracellular antigen levels with that of wild-type AT. The expression experiment did not reveal a significant difference in the antigen levels of the mutant and wild-type AT in the cell lysate, but the mutant AT antigen level was markedly lower than that of its wild-type counterpart in the COS-1 cell supernatant. Immunofluorescence did not indicate any difference between the mutant and wild-type AT in terms of cytoplasmic localization of fluorescence signals. Our findings suggest that the patient's AT deficiency may have been caused by impaired extracellular secretion of mutant AT protein p.Asn87Asp.


Subject(s)
Antigens/metabolism , Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Antithrombin Proteins/genetics , Antithrombin Proteins/physiology , Mutation, Missense , Pregnancy Complications/genetics , Adult , Animals , Antithrombin Proteins/immunology , Antithrombin Proteins/metabolism , COS Cells , Chlorocebus aethiops , Female , Humans , Pregnancy , Thrombophilia/genetics , Venous Thrombosis
16.
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
17.
BMC Vet Res ; 13(1): 182, 2017 Jun 19.
Article in English | MEDLINE | ID: mdl-28629364

ABSTRACT

BACKGROUND: Septic arthritis is a common and potentially devastating disease characterized by severe intra-articular (IA) inflammation and fibrin deposition. Research into equine joint pathologies has focused on inflammation, but recent research in humans suggests that both haemostatic and inflammatory pathways are activated in the joint compartment in arthritic conditions. The aim of this study was to characterize the IA haemostatic and inflammatory responses in horses with experimental lipopolysaccharide (LPS)-induced joint inflammation. Inflammation was induced by IA injection of LPS into one antebrachiocarpal joint of six horses. Horses were evaluated clinically with subjective grading of lameness, and blood and synovial fluid (SF) samples were collected at post injection hours (PIH) -120, -96, -24, 0, 2, 4, 8, 16, 24, 36, 48, 72 and 144. Total protein (TP), white blood cell counts (WBC), serum amyloid A (SAA), haptoglobin, iron, fibrinogen, thrombin-antithrombin (TAT) and d-dimer concentrations were assessed in blood and SF. RESULTS: Intra-articular injection of LPS caused local and systemic signs of inflammation including increased rectal temperature, lameness and increased joint circumference and skin temperature. Most of the biomarkers (TP, WBC, haptoglobin, fibrinogen and TAT) measured in SF increased quickly after LPS injection (at PIH 2-4), whereas SAA and d-dimer levels increased more slowly (at PIH 16 and 144, respectively). SF iron concentrations did not change statistically significantly. Blood WBC, SAA, haptoglobin and fibrinogen increased and iron decreased significantly in response to the IA LPS injection, while TAT and d-dimer concentrations did not change. Repeated pre-injection arthrocenteses caused significant changes in SF concentrations of TP, WBC and haptoglobin. CONCLUSION: Similar to inflammatory joint disease in humans, joint inflammation in horses was accompanied by an IA haemostatic response with changes in fibrinogen, TAT and d-dimer concentrations. Inflammatory and haemostatic responses were induced simultaneously and may likely interact. Further studies of interactions between the two responses are needed for a better understanding of pathogenesis of joint disease in horses. Knowledge of effects of repeated arthrocenteses on levels of SF biomarkers may be of value when markers are used for diagnostic purposes.


Subject(s)
Arthritis, Experimental/veterinary , Biomarkers/metabolism , Horse Diseases/metabolism , Synovial Fluid/metabolism , Animals , Antithrombin Proteins/metabolism , Arthritis, Experimental/blood , Arthritis, Experimental/metabolism , Arthrocentesis/veterinary , Biomarkers/blood , Female , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Hemostasis/drug effects , Horse Diseases/immunology , Horses , Inflammation/metabolism , Injections, Intra-Articular , Lameness, Animal/chemically induced , Lameness, Animal/metabolism , Lipopolysaccharides , Male , Thrombin/metabolism
18.
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
19.
Br J Haematol ; 178(2): 279-285, 2017 07.
Article in English | MEDLINE | ID: mdl-28317092

ABSTRACT

Existing evidence suggests that in most cases antithrombin deficiency can be explained by mutations in its gene, SERPINC1. We investigated the molecular background of antithrombin deficiency in a single centre family cohort study. We included a total of 21 families comprising 15 original probands and sixty-six relatives, 6 of who were surrogate probands for the genetic analysis. Antithrombin activity and antigen levels were measured. The heparin-antithrombin binding ratio assay was used to distinguish between the different subtypes of type II antithrombin deficiency. SERPINC1 mutations were detected by direct sequencing of all 7 exons and regulatory regions, and multiplex ligation-dependent probe amplification. Eighty-six per cent of the families had a detrimental SERPINC1 gene mutation that segregated in the family. We detected 13 different SERPINC1 gene mutations of which 5 were novel. Among all these mutations, 44% was associated with type I deficiency, whereas the remainder was associated with type II heparin binding site (11%), type II pleiotropic effect (33%), type II reactive site (6%) or had the antithrombin Cambridge II mutation (6%). The current study reports several novel SERPINC1 mutations, thereby adding to our knowledge of the molecular background of antithrombin deficiency. Finally, our results point out the importance of future research outside the conventional SERPINC1 gene approach.


Subject(s)
Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Mutation/genetics , Adolescent , Adult , Aged , Antithrombin Proteins/genetics , Child, Preschool , DNA, Recombinant/genetics , Exons/genetics , Female , Humans , Male , Middle Aged , Mutation, Missense/genetics , Pedigree , Young Adult
20.
Menopause ; 24(4): 360-370, 2017 04.
Article in English | MEDLINE | ID: mdl-27922933

ABSTRACT

OBJECTIVE: Vasomotor symptoms (VMS) may be a marker of cardiovascular risk. We aimed to evaluate the cross-sectional association of VMS presence and severity with hemostatic parameter levels measured at baseline among Women's Health Initiative (WHI) Hormone Therapy trial postmenopausal participants. METHODS: This cross-sectional analysis included 2,148 postmenopausal women with measures of VMS presence and severity reported in the 4 weeks before WHI baseline, who were not using warfarin or hormone therapy and for whom the following baseline hemostatic parameters were measured within the WHI Cardiovascular Disease Biomarker Case-Control Study: antithrombin, plasminogen activator inhibitor-1, protein C antigen, total and free protein S antigen, total and free tissue factor pathway inhibitor, D-dimer, normalized activated protein C sensitivity ratio, and thrombin generation. Using multiple linear regression, we estimated the adjusted average difference in each hemostatic parameter associated with VMS presence and severity. A multiple comparisons-corrected P value was computed using the P-min procedure to determine statistical significance of our smallest observed P value. RESULTS: Women were 67 years of age on average and 33% reported VMS presence at baseline. There was some suggestion that VMS presence may be associated with a -0.34 adjusted difference in normalized activated protein C sensitivity ratio compared with no VMS (95% CI, -0.60 to -0.087; P = 0.009), but this association was not significant after correction for multiple comparisons (P = 0.073). VMS presence or severity was not significantly associated with the other hemostatic parameters. CONCLUSIONS: We found no convincing evidence that VMS presence or severity was associated with levels of hemostatic parameters among postmenopausal women.


Subject(s)
Hot Flashes/blood , Postmenopause/blood , Sweating , Aged , Antigens/blood , Antithrombin Proteins/metabolism , Cross-Sectional Studies , Female , Fibrin Fibrinogen Degradation Products/metabolism , Hemostasis , Humans , Lipoproteins/blood , Middle Aged , Plasminogen Activator Inhibitor 1/blood , Protein C/immunology , Protein S/immunology , Severity of Illness Index , Symptom Assessment , Thrombin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...