Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 183
1.
Neurochem Res ; 49(3): 597-616, 2024 Mar.
Article En | MEDLINE | ID: mdl-37978153

Neuroinflammation is assumed as the critical pathophysiologic mechanism of white matter lesions (WMLs), and infiltrated peripheral monocyte-derived macrophages are implicated in the development of neuroinflammation. This study sought to explore the blood molecules that promote the migration of peripheral monocytes to the sites of WMLs. The serum protein expression profiles of patients and Sprague-Dawley rat models with WMLs were detected by data-independent acquisition (DIA) proteomics technique. Compared with corresponding control groups, we acquired 62 and 41 differentially expressed proteins (DEPs) in the serum of patients and model rats with WMLs respectively. Bioinformatics investigations demonstrated that these DEPs were linked to various Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms involved in neuroinflammation. Afterward, we identified thrombin-activatable fibrinolysis inhibitor (TAFI) as a shared and overexpressed protein in clinical and animal serum samples, which was further verified by enzyme-linked immunosorbent assay. Additionally, an upregulation of TAFI was also observed in the white matter of rat models, and the inhibition of TAFI impeded the migration of peripheral monocytes to the area of WMLs. In vitro experiments suggested that TAFI could enhance the migration ability of RAW264.7 cells and increase the expression of Ccr2. Our study demonstrates that neuroinflammatory signals can be detected in the peripheral blood of WMLs patients and model rats. TAFI may serve as a potential protein that promotes the migration of peripheral monocytes to WMLs regions, thereby providing a novel molecular target for further investigation into the interaction between the central and peripheral immune systems.


Carboxypeptidase B2 , White Matter , Humans , Rats , Animals , Fibrinolysis/physiology , Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Neuroinflammatory Diseases , Monocytes/metabolism , Proteomics , White Matter/metabolism , Rats, Sprague-Dawley , Thrombin/metabolism , Thrombin/pharmacology
2.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article En | MEDLINE | ID: mdl-36835137

Carboxypeptidase U (CPU, TAFIa, CPB2) is a potent attenuator of fibrinolysis that is mainly synthesized by the liver as its inactive precursor proCPU. Aside from its antifibrinolytic properties, evidence exists that CPU can modulate inflammation, thereby regulating communication between coagulation and inflammation. Monocytes and macrophages play a central role in inflammation and interact with coagulation mechanisms resulting in thrombus formation. The involvement of CPU and monocytes/macrophages in inflammation and thrombus formation, and a recent hypothesis that proCPU is expressed in monocytes/macrophages, prompted us to investigate human monocytes and macrophages as a potential source of proCPU. CPB2 mRNA expression and the presence of proCPU/CPU protein were studied in THP-1, PMA-stimulated THP-1 cells and primary human monocytes, M-CSF-, IFN-γ/LPS-, and IL-4-stimulated-macrophages by RT-qPCR, Western blotting, enzyme activity measurements, and immunocytochemistry. CPB2 mRNA and proCPU protein were detected in THP-1 and PMA-stimulated THP-1 cells as well as in primary monocytes and macrophages. Moreover, CPU was detected in the cell medium of all investigated cell types and it was demonstrated that proCPU can be activated into functionally active CPU in the in vitro cell culture environment. Comparison of CPB2 mRNA expression and proCPU concentrations in the cell medium between the different cell types provided evidence that CPB2 mRNA expression and proCPU secretion in monocytes and macrophages is related to the degree to which these cells are differentiated. Our results indicate that primary monocytes and macrophages express proCPU. This sheds new light on monocytes and macrophages as local proCPU sources.


Carboxypeptidase B2 , Macrophages , Monocytes , Humans , Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Cell Differentiation/genetics , Inflammation , Macrophage Activation/genetics , Macrophages/metabolism , Monocytes/metabolism , RNA, Messenger
3.
J Thromb Haemost ; 20(5): 1213-1222, 2022 05.
Article En | MEDLINE | ID: mdl-35170225

BACKGROUND: Thrombomodulin on endothelial cells can form a complex with thrombin. This complex has both anticoagulant properties, by activating protein C, and clot-protective properties, by activating thrombin-activatable fibrinolysis inhibitor (TAFI). Activated TAFI (TAFIa) inhibits plasmin-mediated fibrinolysis. OBJECTIVES: TAFIa inhibition is considered a potential antithrombotic strategy. So far, this goal has been pursued by developing compounds that directly inhibit TAFIa. In contrast, we here describe variable domain of heavy-chain-only antibody (VhH) clone 1 that inhibits TAFI activation by targeting human thrombomodulin. METHODS: Two llamas (Lama Glama) were immunized, and phage display was used to select VhH anti-thrombomodulin (TM) clone 1. Affinity was determined with surface plasmon resonance and binding to native TM was confirmed with flow cytometry. Clone 1 was functionally assessed by competition, clot lysis, and thrombin generation assays. Last, the effect of clone 1 on tPA-mediated fibrinolysis in human whole blood was investigated in a microfluidic fibrinolysis model. RESULTS: VhH anti-TM clone 1 bound recombinant TM with a binding affinity of 1.7 ± 0.4 nM and showed binding to native TM. Clone 1 competed with thrombin for binding to TM and attenuated TAFI activation in clot lysis assays and protein C activation in thrombin generation experiments. In a microfluidic fibrinolysis model, inhibition of TM with clone 1 fully prevented TAFI activation. DISCUSSION: We have developed VhH anti-TM clone 1, which inhibits TAFI activation and enhances tPA-mediated fibrinolysis under flow. Different from agents that directly target TAFIa, our strategy should preserve direct TAFI activation via thrombin.


Carboxypeptidase B2 , Carboxypeptidase B2/metabolism , Clone Cells/metabolism , Endothelial Cells/metabolism , Fibrinolysis , Humans , Protein C/metabolism , Thrombin/metabolism , Thrombomodulin/chemistry
4.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article En | MEDLINE | ID: mdl-33916027

Thrombin activatable fibrinolysis inhibitor (TAFI), a proenzyme, is converted to a potent attenuator of the fibrinolytic system upon activation by thrombin, plasmin, or the thrombin/thrombomodulin complex. Since TAFI forms a molecular link between coagulation and fibrinolysis and plays a potential role in venous and arterial thrombotic diseases, much interest has been tied to the development of molecules that antagonize its function. This review aims at providing a general overview on the biochemical properties of TAFI, its (patho)physiologic function, and various strategies to stimulate the fibrinolytic system by interfering with (activated) TAFI functionality.


Carboxypeptidase B2/metabolism , Animals , Carboxypeptidase B2/antagonists & inhibitors , Enzyme Activation , Humans
5.
Laryngoscope ; 131(11): 2413-2420, 2021 11.
Article En | MEDLINE | ID: mdl-33844301

OBJECTIVES/HYPOTHESIS: The objective of this study was to determine the role of thrombin-activatable fibrinolysis inhibitor (TAFI) as a candidate biomarker for therapeutic efficacy of sublingual immunotherapy (SLIT) and to identify the role of TAFI in the pathogenesis of allergic rhinitis (AR). STUDY DESIGN: Retrospective cohort study and laboratory study. METHODS: Serum was collected from patients with allergies to Japanese cedar pollen before, during, and after treatment with SLIT. We measured the levels of immunoreactive TAFI, C3a, and C5a in serum by enzyme-linked immunosorbent assay (ELISA) and assessed their relative impact on a combined symptom-medication score. We also examined the impact of TAFI on mast cells and fibroblasts in experiments performed in vitro. RESULTS: Serum levels of TAFI increased significantly in response to SLIT. By contrast, serum C3a levels decreased significantly over time; we observed a significant negative correlation between serum levels of TAFI versus C3a and symptom-medication score. Mast cell degranulation was inhibited in response to TAFI, as it was the expression of both CCL11 and CCL5 in cultured fibroblasts. CONCLUSIONS: High serum levels of TAFI may be induced by SLIT. TAFI may play a critical protective role in pathogenesis of AR by inactivating C3a and by inhibiting mast cell degranulation and chemokines expression in fibroblasts. LEVEL OF EVIDENCE: 4 Laryngoscope, 131:2413-2420, 2021.


Carboxypeptidase B2/blood , Carboxypeptidase B2/pharmacology , Rhinitis, Allergic/blood , Rhinitis, Allergic/therapy , Sublingual Immunotherapy/methods , Adult , Anaphylatoxins/drug effects , Anaphylatoxins/metabolism , Biomarkers/metabolism , Carboxypeptidase B2/metabolism , Chemokine CCL11/metabolism , Chemokine CCL5/metabolism , Complement C3a/metabolism , Cryptomeria/adverse effects , Cryptomeria/immunology , Enzyme-Linked Immunosorbent Assay/methods , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/pathology , Middle Aged , Retrospective Studies , Rhinitis, Allergic/immunology , Treatment Outcome
6.
J Med Chem ; 64(7): 3897-3910, 2021 04 08.
Article En | MEDLINE | ID: mdl-33764059

Selective and potent inhibitors of activated thrombin activatable fibrinolysis inhibitor (TAFIa) have the potential to increase endogenous and therapeutic fibrinolysis and to behave like profibrinolytic agents without the risk of major hemorrhage, since they do not interfere either with platelet activation or with coagulation during blood hemostasis. Therefore, TAFIa inhibitors could be used in at-risk patients for the treatment, prevention, and secondary prevention of stroke, venous thrombosis, and pulmonary embolisms. In this paper, we describe the design, the structure-activity relationship (SAR), and the synthesis of novel, potent, and selective phosphinanes and azaphosphinanes as TAFIa inhibitors. Several highly active azaphosphinanes display attractive properties suitable for further in vivo efficacy studies in thrombosis models.


Aza Compounds/pharmacology , Carboxypeptidase B2/antagonists & inhibitors , Cyclic P-Oxides/pharmacology , Fibrinolytic Agents/pharmacology , Phosphinic Acids/pharmacology , Protease Inhibitors/pharmacology , Animals , Aza Compounds/chemical synthesis , Aza Compounds/metabolism , Carboxypeptidase B2/metabolism , Catalytic Domain , Cyclic P-Oxides/chemical synthesis , Cyclic P-Oxides/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/metabolism , Humans , Male , Molecular Docking Simulation , Molecular Structure , Phosphinic Acids/chemical synthesis , Phosphinic Acids/metabolism , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
Int J Mol Sci ; 22(2)2021 Jan 17.
Article En | MEDLINE | ID: mdl-33477318

Procarboxypeptidase U (proCPU, TAFI, proCPB2) is a basic carboxypeptidase zymogen that is converted by thrombin(-thrombomodulin) or plasmin into the active carboxypeptidase U (CPU, TAFIa, CPB2), a potent attenuator of fibrinolysis. As CPU forms a molecular link between coagulation and fibrinolysis, the development of CPU inhibitors as profibrinolytic agents constitutes an attractive new concept to improve endogenous fibrinolysis or to increase the efficacy of thrombolytic therapy in thromboembolic diseases. Furthermore, extensive research has been conducted on the in vivo role of CPU in (the acute phase of) thromboembolic disease, as well as on the hypothesis that high proCPU levels and the Thr/Ile325 polymorphism may cause a thrombotic predisposition. In this paper, an overview is given of the methods available for measuring proCPU, CPU, and inactivated CPU (CPUi), together with a summary of the clinical data generated so far, ranging from the current knowledge on proCPU concentrations and polymorphisms as potential thromboembolic risk factors to the positioning of different CPU forms (proCPU, CPU, and CPUi) as diagnostic markers for thromboembolic disease, and the potential benefit of pharmacological inhibition of the CPU pathway.


Carboxypeptidase B2/metabolism , Carboxypeptidase B2/physiology , Thromboembolism/metabolism , Blood Coagulation/physiology , Carboxypeptidase B2/genetics , Fibrinolysin/metabolism , Fibrinolysis/physiology , Genotype , Humans , Thrombin/metabolism , Thromboembolism/physiopathology , Thrombolytic Therapy/methods , Thrombosis/metabolism
8.
Blood Adv ; 4(12): 2631-2639, 2020 06 23.
Article En | MEDLINE | ID: mdl-32556284

Thrombomodulin functions as an anticoagulant through thrombin binding and protein C activation. We herein report the first case of hereditary functional thrombomodulin deficiency presenting with recurrent subcutaneous hemorrhage and old cerebral infarction. The patient had a homozygous substitution of glycine by aspartate at amino acid residue 412 (Gly412Asp) in the thrombin-binding domain of the thrombomodulin gene (designated thrombomodulin-Nagasaki). In vitro assays using a recombinant thrombomodulin with the same mutation as the patient showed a total lack of thrombin binding and activation of protein C and thrombin-activatable fibrinolysis inhibitor (TAFI). Marked clinical and laboratory improvement was obtained with recombinant human soluble thrombomodulin therapy.


Carboxypeptidase B2 , Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Enzyme Activation , Fibrinolysis , Humans , Mutation , Protein C/genetics , Thrombin/metabolism , Thrombomodulin/genetics , Thrombomodulin/metabolism
9.
Eur J Pharmacol ; 880: 173196, 2020 Aug 05.
Article En | MEDLINE | ID: mdl-32416186

Oxaliplatin, a platinum-based chemotherapeutic agent, is widely used to treat colorectal cancer, but it induces peripheral neuropathy as a serious dose-limiting side effect. Recently, thrombomodulin alfa, a recombinant human soluble thrombomodulin, was reported to prevent oxaliplatin-induced peripheral neuropathy in a clinical phase 2 study. Here we conducted preclinical pharmacology studies. Rats were given oxaliplatin (6 mg/kg) intravenously to induce mechanical hyperalgesia associated with peripheral neuropathy. Single intravenous administration of thrombomodulin alfa (0.1, 0.3, 1 mg/kg) dose dependently prevented the development of oxaliplatin-induced mechanical hyperalgesia, with no sex difference in the efficacy. The preventative effect of thrombomodulin alfa on mechanical hyperalgesia was attenuated by antithrombin or carboxypeptidase inhibitor. In addition, carboxypeptidase B, a homolog of activated thrombin-activatable fibrinolysis inhibitor (TAFI) and human-derived activated protein C, prevented mechanical hyperalgesia, whereas antithrombin or other anti-coagulants did not. These results suggest that thrombomodulin alfa prevents sensory symptoms of oxaliplatin-induced peripheral neuropathy through the activation of TAFI and protein C by modulating thrombin activity, but the effects are independent of an anticoagulant effect. On the other hand, thrombomodulin alfa did not affect the anti-cancer activity of oxaliplatin on human colon cancer cell lines or mice transplanted with HCT116 cells. These results indicate that thrombomodulin alfa prevents sensory symptoms of oxaliplatin-induced peripheral neuropathy without affecting the anti-tumor activity of oxaliplatin. Therefore, thrombomodulin alfa is a promising drug to prevent the symptoms of oxaliplatin-induced peripheral neuropathy.


Analgesics/therapeutic use , Antineoplastic Agents/adverse effects , Colonic Neoplasms/drug therapy , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/drug therapy , Thrombomodulin/therapeutic use , Analgesics/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carboxypeptidase B2/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Female , Humans , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Neuralgia/chemically induced , Neuralgia/metabolism , Oxaliplatin/therapeutic use , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Protein C/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombomodulin/genetics , Touch
10.
Haemophilia ; 26(4): e151-e160, 2020 Jul.
Article En | MEDLINE | ID: mdl-32325538

INTRODUCTION: Plasma-derived FVIII/VWF complex was reported to be less sensitive to inhibitors than FVIII preparations devoid of VWF. AIM: To compare the efficacy of FVIII/VWF complex (Fanhdi) and five different VWF-free FVIII preparations in restoring thrombin generation and activation of thrombin-activatable fibrinolysis inhibitor (TAFI) in haemophilic plasma, with and without inhibitor, and in cell-based models. METHODS: Experiments were performed in haemophilic plasma supplemented with inhibitory IgG or in plasma samples obtained from haemophilia A patients without (n = 11) and with inhibitor (n = 12). Thrombin generation was evaluated by calibrated automated thrombography (CAT) under standard conditions, in the presence of activated protein C (APC) or thrombomodulin (TM), and in cell-based models including endothelial cells, either alone or in combination with platelets or tissue factor-expressing blood mononuclear cells. The kinetics of TAFI activation was determined by a two-stage functional assay in the absence and in the presence of APC. RESULTS: In haemophilic plasma without inhibitor, Fanhdi enhanced thrombin generation and TAFI activation as well as recombinant (2nd-4th generation) and plasma-derived FVIII preparations devoid of VWF. On the contrary, in plasma with inhibitor, Fanhdi displayed a greater ability to restore thrombin generation and TAFI activation under all tested conditions. Notably, in cell-based models including endothelial cells, Fanhdi proved more efficient than all other preparations in improving thrombin generation even in the absence of inhibitor. CONCLUSION: The greater pro-haemostatic activity of FVIII/VWF complex, either in haemophilic plasma with inhibitor or in the presence of endothelial cells, may offer therapeutic advantages.


Factor VIII/pharmacology , Hemophilia A/drug therapy , von Willebrand Factor/pharmacology , Carboxypeptidase B2/drug effects , Carboxypeptidase B2/metabolism , Carboxypeptidase B2/pharmacology , Coagulants/pharmacology , Coagulants/therapeutic use , Combined Modality Therapy , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Factor VIII/therapeutic use , Fibrinolysis/drug effects , Hemophilia A/blood , Hemostasis/drug effects , Hemostasis/physiology , Humans , Immunoglobulin G/metabolism , Kinetics , Plasma/metabolism , Protein C/metabolism , Thrombin/drug effects , Thrombin/metabolism , Thrombomodulin/metabolism , Thromboplastin/metabolism , Treatment Outcome , von Willebrand Factor/therapeutic use
11.
Am J Cardiol ; 125(5): 751-758, 2020 03 01.
Article En | MEDLINE | ID: mdl-31889526

Recent findings in atrial fibrillation (AF) patients receiving oral anticoagulation showed that diabetes without insulin therapy has a thromboembolic risk comparable to nondiabetic patients, whereas only diabetic patients on insulin have a heightened thromboembolic risk. We explored possible pathophysiological correlates of such finding on 90 AF patients on oral anticoagulation, divided according to diabetes status (n = 30 without diabetes; n = 29 with diabetes on oral antidiabetic drugs; n = 31 with insulin-requiring diabetes). We assessed von Willebrand Factor (VWF) concentration (VWF:Ag) and activity (VWF R:Co) as measures of endothelial dysfunction; and thrombin-activatable fibrinolysis inhibitor (TAFI) and prothrombin fragment 1 + 2 (F1+2) levels as markers of fibrinolytic activity and thrombin generation. Values of VWF:Ag, VWF:RCo, and TAFI were similar in the 3 groups. Patients with diabetes requiring insulin had significantly higher levels of F1+2 (median 23.1 pg/ml [interquartile range 17.6; 33.5]) than those without diabetes (16.3 pg/ml [11.5; 22.5], p = 0.036) and diabetic patients on oral antidiabetic drugs (20.6 pg/ml [13.3; 29], p = 0.046). Thus, in AF patients receiving oral anticoagulation, those with diabetes, regardless of the diabetes type (with or without insulin therapy), and those without diabetes have comparable indices of the explored parameters of endothelial dysfunction and fibrinolytic activity. Despite anticoagulant therapy, thrombin generation is selectively higher in diabetic patients' on insulin than in those without diabetes or with diabetes on oral antidiabetic drugs, with no differences between these latter 2 conditions. Thrombin generation might thus be a predominant contributor to the excess of thromboembolic risk in AF patients on insulin-requiring diabetes.


Atrial Fibrillation/metabolism , Carboxypeptidase B2/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Peptide Fragments/metabolism , Prothrombin/metabolism , von Willebrand Factor/metabolism , Aged , Aged, 80 and over , Anticoagulants/therapeutic use , Antithrombins/therapeutic use , Atrial Fibrillation/drug therapy , Atrial Fibrillation/epidemiology , Case-Control Studies , Comorbidity , Cross-Sectional Studies , Dabigatran/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Endothelium, Vascular/physiopathology , Factor Xa Inhibitors/therapeutic use , Female , Humans , Italy/epidemiology , Male , Metformin/therapeutic use , Sulfonylurea Compounds/therapeutic use
12.
JCI Insight ; 4(19)2019 10 03.
Article En | MEDLINE | ID: mdl-31465300

Excessive vascular remodeling is characteristic of hemophilic arthropathy (HA) and may contribute to joint bleeding and the progression of HA. Mechanisms for pathological vascular remodeling after hemophilic joint bleeding are unknown. In hemophilia, activation of thrombin-activatable fibrinolysis inhibitor (TAFI) is impaired, which contributes to joint bleeding and may also underlie the aberrant vascular remodeling. Here, hemophilia A (factor VIII-deficient; FVIII-deficient) mice or TAFI-deficient mice with transient (antibody-induced) hemophilia A were used to determine the role of FVIII and TAFI in vascular remodeling after joint bleeding. Excessive vascular remodeling and vessel enlargement persisted in FVIII-deficient and TAFI-deficient mice, but not in transient hemophilia WT mice, after similar joint bleeding. TAFI-overexpression in FVIII-deficient mice prevented abnormal vessel enlargement and vascular leakage. Age-related vascular changes were observed with FVIII or TAFI deficiency and correlated positively with bleeding severity after injury, supporting increased vascularity as a major contributor to joint bleeding. Antibody-mediated inhibition of uPA also prevented abnormal vascular remodeling, suggesting that TAFI's protective effects include inhibition of uPA-mediated plasminogen activation. In conclusion, the functional TAFI deficiency in hemophilia drives maladaptive vascular remodeling in the joints after bleeding. These mechanistic insights allow targeted development of potentially new strategies to normalize vascularity and control rebleeding in HA.


Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Factor VIII/genetics , Hemarthrosis/complications , Hemophilia A/complications , Hemophilia A/genetics , Vascular Remodeling/physiology , Animals , Disease Models, Animal , Factor VIII/metabolism , Female , Genetic Predisposition to Disease/genetics , Hemarthrosis/pathology , Hemophilia A/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transcriptome
13.
J Med Entomol ; 56(1): 72-85, 2019 01 08.
Article En | MEDLINE | ID: mdl-30124910

Malaria is a vector-borne infectious disease that is considered a priority of the World Health Organization due to its enormous impacts on global health. Plasmodium spp. (Haemosporida: Plasmodiidae), Anopheles spp. (Diptera: Culicidae), and a suitable host are the key elements for malaria transmission. To disrupt the parasitic life cycle of malaria or prevent its transmission, these three key elements should be targeted by effective control strategies. Development of vaccines that interrupt malaria transmission is one of the solutions that has been recommended to the countries that aim to eliminate malaria. With respect to the important role of Anopheles stephensi in malaria transmission and involvement of Anopheles carboxypeptidase B1 in sexual parasite development, we characterized the second member of cpb gene family (cpbAs2) of An. Stephensi to provide some basic information and evaluate significance of cpbAs2's role in complementing sexual plasmodium development role of cpbAs1. The cpbAs2 mRNA sequence was characterized by 3' and 5' RACE and the structural features of its coded protein were studied by in silico modeling. The coding sequence and gene structure of cpbAs2 were determined empirically and compared with the in silico predictions from the An. stephensi genome sequencing project. Furthermore, homology modeling revealed that its structure is very similar to the structurally important domains of procarboxypeptidase B2 in humans. This study provides basic molecular and structural information about another member of the cpb gene family of An. stephensi. The reported results are informative and necessary for evaluation of the role of this gene in sexual parasite development by future studies.


Anopheles/enzymology , Carboxypeptidase B2/genetics , Amino Acid Sequence , Animals , Anopheles/parasitology , Base Sequence , Carboxypeptidase B2/chemistry , Carboxypeptidase B2/metabolism , Computer Simulation , Female , Glycosylation , Plasmodium/growth & development , Sequence Analysis, DNA , Structural Homology, Protein
14.
Blood ; 132(15): 1593-1603, 2018 10 11.
Article En | MEDLINE | ID: mdl-30026184

Joint bleeds are common in congenital hemophilia but rare in acquired hemophilia A (aHA) for reasons unknown. To identify key mechanisms responsible for joint-specific bleeding in congenital hemophilia, bleeding phenotypes after joint injury and tail transection were compared in aHA wild-type (WT) mice (receiving an anti-factor VIII [FVIII] antibody) and congenital HA (FVIII-/-) mice. Both aHA and FVIII-/- mice bled severely after tail transection, but consistent with clinical findings, joint bleeding was notably milder in aHA compared with FVIII-/- mice. Focus was directed to thrombin-activatable fibrinolysis inhibitor (TAFI) to determine its potentially protective effect on joint bleeding in aHA. Joint bleeding in TAFI-/- mice with anti-FVIII antibody was increased, compared with WT aHA mice, and became indistinguishable from joint bleeding in FVIII-/- mice. Measurements of circulating TAFI zymogen consumption after joint injury indicated severely defective TAFI activation in FVIII-/- mice in vivo, consistent with previous in vitro analyses in FVIII-deficient plasma. In contrast, notable TAFI activation was observed in aHA mice, suggesting that TAFI protected aHA joints against bleeding. Pharmacological inhibitors of fibrinolysis revealed that urokinase-type plasminogen activator (uPA)-induced fibrinolysis drove joint bleeding, whereas tissue-type plasminogen activator-mediated fibrinolysis contributed to tail bleeding. These data identify TAFI as an important modifier of hemophilic joint bleeding in aHA by inhibiting uPA-mediated fibrinolysis. Moreover, our data suggest that bleed protection by TAFI was absent in congenital FVIII-/- mice because of severely defective TAFI activation, underscoring the importance of clot protection in addition to clot formation when considering prohemostatic strategies for hemophilic joint bleeding.


Carboxypeptidase B2/metabolism , Hemarthrosis/etiology , Hemarthrosis/metabolism , Hemophilia A/complications , Animals , Carboxypeptidase B2/genetics , Disease Models, Animal , Gene Deletion , Hemarthrosis/genetics , Hemophilia A/genetics , Hemophilia A/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Urokinase-Type Plasminogen Activator/metabolism
16.
Sci Rep ; 8(1): 505, 2018 01 11.
Article En | MEDLINE | ID: mdl-29323190

Angiopoietin-1 (Ang1) and Angiopoietin-2 (Ang2) are ligands for Tie2, an endothelial-specific receptor tyrosine kinase that is an essential regulator of angiogenesis. Here we report the identification, via expression cloning, of thrombomodulin (TM) as another receptor for Ang1 and Ang2. Thrombomodulin is an endothelial cell surface molecule that plays an essential role as a coagulation inhibitor via its function as a cofactor in the thrombin-mediated activation of protein C, an anticoagulant protein, as well as thrombin-activatable fibrinolysis inhibitor (TAFI). Ang1 and Ang2 inhibited the thrombin/TM-mediated generation of activated protein C and TAFI in cultured endothelial cells, and inhibited the binding of thrombin to TM in vitro. Ang2 appears to bind TM with higher affinity than Ang1 and is a more potent inhibitor of TM function. Consistent with a potential role for angiopoietins in coagulation, administration of thrombin to mice rapidly increased plasma Ang1 levels, presumably reflecting release from activated platelets (previously shown to contain high levels of Ang1). In addition, Ang1 levels were significantly elevated in plasma prepared from wound blood, suggesting that Ang1 is released from activated platelets at sites of vessel injury. Our results imply a previously undescribed role for angiopoietins in the regulation of hemostasis.


Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Thrombin/metabolism , Thrombomodulin/metabolism , Angiopoietin-1/blood , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , COS Cells , Carboxypeptidase B2/metabolism , Chlorocebus aethiops , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Platelet Factor 4/metabolism , Protein Binding , Protein C/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Thrombin/chemistry , Thrombin/pharmacology , Thrombomodulin/genetics
17.
Dig Dis ; 36(3): 244-251, 2018.
Article En | MEDLINE | ID: mdl-29332096

AIM: Coagulation disorders may develop in association with severe acute pancreatitis (AP). Plasma thrombin-antithrombin III complex (TAT) levels are one of the principal markers of coagulation disorder. The purpose of this study was to evaluate TAT and other hemostatic parameters in patients with AP and to examine whether or not these parameters indicate the severity of AP. METHOD: Forty-six patients with AP (14 severe, 32 non-severe) and a 30-member healthy control group were recruited. The severity of AP was determined using the revised Atlanta classification. ELISA was used to measure patients' plasma TAT levels. RESULTS: The TAT levels of AP patients at presentation were higher than those of the control group (p = 0.005). The plasma TAT levels of patients with severe AP were also significantly higher than those of patients with non-severe AP (p = 0.05) and of the control group (p < 0.001). The general accuracy, sensitivity and specificity of TAT levels in predicting the severity of AP were 77.4, 77.8, and 77.3% respectively. CONCLUSION: The coagulation cascade was activated in the AP patients in our study, and this was shown to become more pronounced as severity of the disease increased. Plasma TAT levels at the time of presentation in patients with AP can be used as a marker for predicting the severity of the disease.


Antithrombin III/metabolism , Pancreatitis/blood , Peptide Hydrolases/metabolism , Acute Disease , Biomarkers/blood , C-Reactive Protein/metabolism , Carboxypeptidase B2/metabolism , Case-Control Studies , Female , Humans , Male , Middle Aged , ROC Curve
18.
Thromb Haemost ; 117(8): 1509-1517, 2017 07 26.
Article En | MEDLINE | ID: mdl-28640323

The thrombin-thrombomodulin (TM) complex activates thrombin-activable fibrinolysis inhibitor (TAFI) more efficiently than thrombin alone. The exosite on TAFI required for its TM-dependent activation by thrombin has not been identified. Based on previous work by us and others, we generated TAFI variants with one or more of residues Lys 42, Lys 43, Lys 44 and Arg 12 within the activation peptide mutated to alanine. Mutation of one, two, or three Lys residues or the Arg residue alone decreased the catalytic efficiency of TAFI activation by thrombin-TM by 2.4-, 3.2-, 4.7-, and 15.0-fold, respectively, and increased the TAFI concentrations required for half-maximal prolongation of clot lysis times (K1/2) by 3-, 4,- 15-, and 24-fold, respectively. Mutation of all four residues decreased the catalytic efficiency of TAFI activation by 45.0-fold, increased the K1/2 by 130-fold, and abolished antifibrinolytic activity in a clot lysis assay at physiologic levels of TAFI. Similar trends in the antifibrinolytic activity of the TAFI variants were observed when plasma clots were formed using HUVECs as the source of TM. When thrombin was used as the activator, mutation of all four residues reduced the rate of activation by 1.1-fold compared with wild-type TAFI, suggesting that these mutations only impacted activation kinetics in the presence of TM. Surface plasmon resonance data suggest that mutation of the four residues abrogates TM binding with or without thrombin. Therefore, Lys 42, Lys 43, Lys 44 and Arg 12 are critical for the interaction of TAFI with the thrombin-TM complex, which modulates its antifibrinolytic potential.


Carboxypeptidase B2/metabolism , Fibrinolysis , Thrombomodulin/blood , Animals , Arginine , Carboxypeptidase B2/genetics , Cricetinae , Enzyme Activation , Fibrinolysis/genetics , Genotype , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kinetics , Lysine , Mutation , Phenotype , Protein Binding , Thrombin/metabolism
19.
PLoS One ; 12(5): e0177117, 2017.
Article En | MEDLINE | ID: mdl-28472123

OBJECTIVE: Thrombin-activatable fibrinolysis inhibitor (TAFI) reduces the breakdown of fibrin clots through its action as an indirect inhibitor of plasmin. Studies in TAFI-deficient mice have implicated a potential role for TAFI in Abdominal Aortic Aneurysm (AAA) disease. The role of TAFI inhibition on AAA formation in adult ApoE-/- mice is unknown. The aim of this paper was to investigate the effects of TAFI inhibition on AAA development and progression. METHODS: Using the Angiotensin II model of AAA, male ApoE-/- mice were infused with Angiotensin II 750ng/kg/min with or without a monoclonal antibody inhibitor of plasmin-mediated activation of TAFI, MA-TCK26D6, or a competitive small molecule inhibitor of TAFI, UK-396082. RESULTS: Inhibition of TAFI in the Angiotensin II model resulted in a decrease in the mortality associated with AAA rupture (from 40.0% to 16.6% with MA-TCK26D6 (log-rank Mantel Cox test p = 0.16), and 8.3% with UK-396082 (log-rank Mantel Cox test p = 0.05)). Inhibition of plasmin-mediated TAFI activation reduced the incidence of AAA from 52.4% to 30.0%. However, late treatment with MA-TCK26D6 once AAA were already established had no effect on the progression of AAA in this model. CONCLUSIONS: The formation of intra-mural thrombus is responsible for the dissection and early rupture in the angiotensin II model of AAA, and this process can be prevented through inhibition of TAFI. Late treatment with a TAFI inhibitor does not prevent AAA progression. These data may indicate a role for inhibition of plasmin-mediated TAFI activation in the early stages of AAA development, but not in its progression.


Carboxypeptidase B2/antagonists & inhibitors , Disease Models, Animal , Fibrinolysin/metabolism , Animals , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/genetics , Carboxypeptidase B2/metabolism , Disease Progression , Male , Mice , Mice, Knockout
20.
Circ Res ; 120(8): 1246-1262, 2017 Apr 14.
Article En | MEDLINE | ID: mdl-28289017

RATIONALE: Pulmonary hypertension is a fatal disease; however, its pathogenesis still remains to be elucidated. Thrombin-activatable fibrinolysis inhibitor (TAFI) is synthesized by the liver and inhibits fibrinolysis. Plasma TAFI levels are significantly increased in chronic thromboembolic pulmonary hypertension (CTEPH) patients. OBJECTIVE: To determine the role of activated TAFI (TAFIa) in the development of CTEPH. METHODS AND RESULTS: Immunostaining showed that TAFI and its binding partner thrombomodulin (TM) were highly expressed in the pulmonary arteries (PAs) and thrombus in patients with CTEPH. Moreover, plasma levels of TAFIa were increased 10-fold in CTEPH patients compared with controls. In mice, chronic hypoxia caused a 25-fold increase in plasma levels of TAFIa with increased plasma levels of thrombin and TM, which led to thrombus formation in PA, vascular remodeling, and pulmonary hypertension. Consistently, plasma clot lysis time was positively correlated with plasma TAFIa levels in mice. Additionally, overexpression of TAFIa caused organized thrombus with multiple obstruction of PA flow and reduced survival rate under hypoxia in mice. Bone marrow transplantation showed that circulating plasma TAFI from the liver, not in the bone marrow, was activated locally in PA endothelial cells through interactions with thrombin and TM. Mechanistic experiments demonstrated that TAFIa increased PA endothelial permeability, smooth muscle cell proliferation, and monocyte/macrophage activation. Importantly, TAFIa inhibitor and peroxisome proliferator-activated receptor-α agonists significantly reduced TAFIa and ameliorated animal models of pulmonary hypertension in mice and rats. CONCLUSIONS: These results indicate that TAFIa could be a novel biomarker and realistic therapeutic target of CTEPH.


Arterial Pressure , Carboxypeptidase B2/metabolism , Hypertension, Pulmonary/etiology , Liver/metabolism , Pulmonary Artery/metabolism , Thromboembolism/complications , Adult , Animals , Capillary Permeability , Carboxypeptidase B2/deficiency , Carboxypeptidase B2/genetics , Case-Control Studies , Cell Proliferation , Chronic Disease , Disease Models, Animal , Female , Hep G2 Cells , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Liver/drug effects , Macrophage Activation , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , PPAR alpha/agonists , PPAR alpha/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Pyrimidines/pharmacology , Rats, Sprague-Dawley , Signal Transduction , Thrombin/metabolism , Thromboembolism/metabolism , Thromboembolism/physiopathology , Thromboembolism/prevention & control , Thrombomodulin/metabolism , Transfection , Up-Regulation
...