Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Thromb Res ; 210: 78-86, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35030422

ABSTRACT

INTRODUCTION: Thrombolysis using recombinant tissue-type plasminogen activator (rt-PA) is the pharmacological treatment of choice in acute thrombotic events. However, a narrow therapeutic window and bleeding complications limit its use. We describe the role of carboxypeptidase inhibitor from potato tuber (PTCI), an inhibitor of activated thrombin-activatable fibrinolysis inhibitor (TAFIa), on Glu-plasminogen accumulation and microthrombus dynamics in vivo and demonstrate its influence on rt-PA-mediated thrombolysis. MATERIALS AND METHODS: In conjunction with real-time intravital two-photon excitation fluorescence microscopy, we produced and imaged laser-induced microthrombi in the mesenteric venules of Green Fluorescent Protein (GFP)-expressing mice. We examined microthrombus dynamics and thrombolysis patterns in vivo by measuring the changes in the fluorescence intensity of labeled Glu-plasminogen following administration of epsilon aminocaproic acid (EACA), PTCI, and rt-PA. RESULTS: PTCI enhanced Glu-plasminogen accumulation at the core of the thrombus by inhibiting TAFIa, while EACA inhibited this process. Exogenous rt-PA effectively triggered Glu-plasminogen activation within the thrombus and promoted thrombolysis. Administration of PTCI and rt-PA together showed no significant benefit on thrombolysis compared to rt-PA administration alone. However, early-phase systemic administration of PTCI before thrombolytic therapy by rt-PA expedited clot lysis as evidenced by significantly faster time to reach peak Glu-plasminogen fluorescence intensity and shorter time to achieve near-complete clot lysis (P = 0.014 and P = 0.003, respectively). CONCLUSIONS: PTCI potentiates rt-PA-mediated thrombolysis when administered early in acute thrombotic events. Further studies are warranted to explore the potential of TAFI inhibitors as adjunct agents in thrombolysis or thromboprophylaxis.


Subject(s)
Carboxypeptidases/antagonists & inhibitors , Thrombosis , Venous Thromboembolism , Animals , Anticoagulants/therapeutic use , Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis , Humans , Intravital Microscopy , Mice , Thrombolytic Therapy , Thrombosis/drug therapy , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use , Venous Thromboembolism/drug therapy
2.
Thromb Res ; 204: 81-87, 2021 08.
Article in English | MEDLINE | ID: mdl-34153648

ABSTRACT

Enhancement of fibrinolysis constitutes a promising approach to treat thrombotic diseases. Venous thrombosis and thromboembolism risks are associated with increased plasma levels of TAFI (Thrombin Activatable Fibrinolysis Inhibitor) as well as its active form TAFIa. A new TAFIa inhibitor, namely S62798 has been identified. Its ability to enhance fibrinolysis was investigated both in vitro and in vivo in a mouse model of pulmonary thromboembolism, as well as its effect on bleeding. S62798 is a highly selective human, mouse and rat TAFIa inhibitor (IC50 = 11; 270; 178 nmol/L, respectively). It accelerates lysis of a human clot in vitro, evaluated by thromboelastometry (EC50 = 27 nmol/L). In a rat tail bleeding model, no effect of S62798 treatment was observed up to 20 mg/kg. Enhancement of endogenous fibrinolysis by S62798 was investigated in a mouse model of Tissue Factor-induced pulmonary thromboembolism. Intravenous administration of S62798 decreased pulmonary fibrin clots with a minimal effective dose of 0.03 mg/kg. Finally, effect of S62798 in combination with heparin was evaluated. When treatment of heparin was done in a curative setting, no effect was observed whereas a significantly decreased pulmonary fibrin deposition was observed in response to S62798 alone or in combination with heparin. This study demonstrates that S62798 is a potent TAFIa inhibitor with minimal risk of bleeding. In vivo, curative S62798 intravenous treatment, alone or associated with heparin, accelerated clot lysis by potentiating endogenous fibrinolysis and thus decreased pulmonary fibrin clots. S62798 is expected to be a therapeutic option for pulmonary embolism patients on top of anticoagulants.


Subject(s)
Carboxypeptidase B2 , Enzyme Inhibitors/pharmacology , Pulmonary Embolism , Animals , Carboxypeptidase B2/antagonists & inhibitors , Disease Models, Animal , Fibrin Clot Lysis Time , Fibrinolysis , Humans , Mice , Pulmonary Embolism/drug therapy , Rats
3.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916027

ABSTRACT

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.


Subject(s)
Carboxypeptidase B2/metabolism , Animals , Carboxypeptidase B2/antagonists & inhibitors , Enzyme Activation , Humans
4.
J Med Chem ; 64(7): 3897-3910, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33764059

ABSTRACT

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.


Subject(s)
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
5.
J Thromb Haemost ; 17(6): 878-884, 2019 06.
Article in English | MEDLINE | ID: mdl-30887647

ABSTRACT

Essentials Hemolytic influence on the (pro)carboxypeptidase U ((pro)CPU) system is not known. In the current manuscript, this was assessed by spiking pooled normal plasma with hemolysate. CPU activity, proCPU levels, and clot lysis times showed a dose-dependent hemolytic bias. The observed bias in the several CPU related parameters is due to inhibition of CPU activity. INTRODUCTION: Spurious hemolysis of samples is the leading cause of interference in coagulation testing and was described to interfere in fibrinolysis assays. The influence of hemolysis on the procarboxypeptidase U (proCPU) system is not known. METHODS: By means of spiking of hemolysate in pooled normal plasma, the effect of hemolysis on CPU, proCPU, and functional clot lysis assays was assessed. The influence of hemolysis on CPU generation during in vitro clot lysis was also evaluated. Cutoffs corresponding to maximal acceptable bias were determined. RESULTS AND DISCUSSION: When active CPU was added to pooled plasma, a severe decrease in activity - up to 97.2% inhibition - was seen with increasing plasma concentrations of oxyhemoglobin (oxyHb) and the 10% cutoff value was found to be 0.3 g/L oxyHb. Using an activity-based assay, proCPU levels appeared to decrease gradually with increased hemolysis (maximal reduction of 19.5%) with a 10% cutoff value of 4.2 g/L oxyHb. The relative clot lysis time (CLT) showed a maximal negative bias of 68.5%. The reduction in CLT paralleled a significant reduction of the first CPU activity peak during clot lysis. The cutoff value for the CLT was 0.4 g/L oxyHb. In presence of thrombomodulin (TM), CLT+TM was not affected up to 8.0 g/L oxyHb. CONCLUSION: These data indicate a clear inhibition of the CPU system because of hemolysis resulting in an increase of lysis in functional fibrinolysis assays. We were able to quantify the inhibitory effect and to propose cutoff values for every parameter.


Subject(s)
Blood Coagulation Tests/methods , Carboxypeptidase B2/antagonists & inhibitors , Carboxypeptidase B2/blood , Hemolysis/physiology , Blood Coagulation Tests/statistics & numerical data , Fibrin Clot Lysis Time/methods , Fibrin Clot Lysis Time/statistics & numerical data , Fibrinolysis/physiology , Healthy Volunteers , Humans , In Vitro Techniques
6.
J Thromb Haemost ; 16(10): 2057-2069, 2018 10.
Article in English | MEDLINE | ID: mdl-30053349

ABSTRACT

Essentials AZD9684 is a potent inhibitor of carboxypeptidase U (CPU, TAFIa, CPB2). The effect of AZD9684 on fibrinolysis was investigated in four in vitro systems. The CPU system also attenuates fibrinolysis in more advanced hemostatic systems. The size of the observed effect on fibrinolysis is dependent on the exact experimental conditions. SUMMARY: Background Carboxypeptidase U (CPU, carboxypeptidase B2, activated thrombin-activatable fibrinolysis inhibitor) is a basic carboxypeptidase that attenuates fibrinolysis. This characteristic has raised interest in the scientific community and pharmaceutical industry for the development of inhibitors as profibrinolytic agents. Objectives Little is known about the contribution of CPU to clot resistance in more advanced hemostatic models, which include blood cells and shear stress. The aim of this study was to evaluate the effects of the CPU system in in vitro systems for fibrinolysis with different grades of complexity. Methods The contribution of the CPU system was evaluated in the following systems: (i) plasma clot lysis; (ii) rotational thromboelastometry (ROTEM) in whole blood; (iii) front lysis with confocal microscopy in platelet-free and platelet-rich plasma; and (iv) a microfluidic system with whole blood under arterial shear stress. Experiments were carried out in the presence or absence of AZD9684, a specific CPU inhibitor. Results During plasma clot lysis, addition of AZD9684 resulted in 33% faster lysis. In ROTEM, the lysis onset time was decreased by 38%. For both clot lysis and ROTEM, an AZD9684 dose-dependent response was observed. CPU inhibition in front lysis experiments resulted in 47% and 50% faster lysis for platelet-free plasma and platelet-rich plasma, respectively. Finally, a tendency for faster lysis was observed only in the microfluidic system when AZD9684 was added. Conclusions Overall, these experiments provide novel evidence that the CPU system can also modulate fibrinolysis in more advanced hemostatic systems. The extent of the effects appears to be dependent upon the exact experimental conditions.


Subject(s)
Blood Coagulation Tests/methods , Butyrates/pharmacology , Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Protease Inhibitors/pharmacology , Pyridines/pharmacology , Carboxypeptidase B2/blood , Humans , Kinetics
7.
Bioorg Med Chem Lett ; 28(13): 2256-2260, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29859906

ABSTRACT

Activated thrombin-activatable fibrinolysis inhibitor (TAFIa) is a target molecule for treating thromboembolic disorders. We previously reported that design and synthesis of compound 1 containing a selenol group and chloloaminopyridine. Compound 1 showed high inhibitory activity towards TAFIa, with a high degree of selectivity for TAFIa over carboxypeptidase N (CPN). Here we report investigation of this selectivity. To obtain co-crystal of 1/pp-CPB (a surrogate of TAFIa), we synthesized protected compound 5 as a stabilized precursor of 1. The X-ray crystal structure and docking study indicated that the Cl substituent is accommodated in the pp-CPB specific pocket whereas CPN has no identical pocket. This is important information for the design of drugs targeting TAFIa with high selectivity.


Subject(s)
Aminopyridines/chemistry , Carboxypeptidase B2/antagonists & inhibitors , Organoselenium Compounds/chemistry , Protease Inhibitors/chemistry , Aminopyridines/chemical synthesis , Animals , Binding Sites , Carboxypeptidase B/antagonists & inhibitors , Carboxypeptidase B/chemistry , Humans , Hydrogen Bonding , Lysine Carboxypeptidase/antagonists & inhibitors , Molecular Docking Simulation , Organoselenium Compounds/chemical synthesis , Protease Inhibitors/chemical synthesis , Swine
8.
PLoS One ; 12(5): e0177117, 2017.
Article in English | MEDLINE | ID: mdl-28472123

ABSTRACT

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.


Subject(s)
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
9.
Thromb Haemost ; 117(4): 682-690, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28150854

ABSTRACT

Using intravital confocal microscopy, we observed previously that the process of platelet phosphatidylserine (PS) exposure, fibrin formation and lysine binding site-dependent plasminogen (plg) accumulation took place only in the centre of thrombi, not at their periphery. These findings prompted us to analyse the spatiotemporal regulatory mechanisms underlying coagulation and fibrinolysis. We analysed the fibrin network formation and the subsequent lysis in an in vitro experiment using diluted platelet-rich plasma supplemented with fluorescently labelled coagulation and fibrinolytic factors, using confocal laser scanning microscopy. The structure of the fibrin network formed by supplemented tissue factor was uneven and denser at the sites of coagulation initiation regions (CIRs) on PS-exposed platelets. When tissue-type plasminogen activator (tPA; 7.5 nM) was supplemented, labelled plg (50 nM) as well as tPA accumulated at CIRs, from where fibrinolysis started and gradually expanded to the peripheries. The lysis time at CIRs and their peripheries (50 µm from the CIR) were 27.9 ± 6.6 and 44.4 ± 9.7 minutes (mean ± SD, n=50 from five independent experiments) after the addition of tissue factor, respectively. Recombinant human soluble thrombomodulin (TMα; 2.0 nM) attenuated the CIR-dependent plg accumulation and strongly delayed fibrinolysis at CIRs. A carboxypeptidase inhibitor dose-dependently enhanced the CIR-dependent fibrinolysis initiation, and at 20 µM it completely abrogated the TMα-induced delay of fibrinolysis. Our findings are the first to directly present crosstalk between coagulation and fibrinolysis, which takes place on activated platelets' surface and is further controlled by thrombin-activatable fibrinolysis inhibitor (TAFI).


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/drug effects , Carboxypeptidase B2/pharmacology , Fibrinolysis/drug effects , Intravital Microscopy/methods , Microscopy, Confocal/methods , Platelet Activation/drug effects , Platelet-Rich Plasma/diagnostic imaging , Blood Platelets/enzymology , Carboxypeptidase B2/antagonists & inhibitors , Dose-Response Relationship, Drug , Fibrin/metabolism , Humans , Phosphatidylserines/metabolism , Platelet-Rich Plasma/cytology , Platelet-Rich Plasma/enzymology , Protease Inhibitors/pharmacology , Thrombomodulin/metabolism , Time Factors
10.
J Thromb Haemost ; 15(5): 961-971, 2017 05.
Article in English | MEDLINE | ID: mdl-28211169

ABSTRACT

Essentials DS-1040 inhibits the activated form of thrombin-activatable fibrinolysis inhibitor (TAFIa). Infusion of DS-1040 was safe and well tolerated in healthy young and elderly subjects. DS-1040 substantially decreased TAFIa activity but had no impact on bleeding time. DS-1040 may provide an option of safer thrombolytic therapy. SUMMARY: Background Current treatments for acute ischemic stroke and venous thromboembolism, such as recombinant tissue-type plasminogen activator and thrombectomy, are limited by a narrow time window and the risk of bleeding. DS-1040 is a novel low molecular weight compound that inhibits the activated form of thrombin-activatable fibrinolysis inhibitor (TAFIa), and was developed as a fibrinolysis enhancer for the treatment of thromboembolic diseases. Objectives This first-in-human, randomized, placebo-controlled, three-part, phase 1 study was conducted to evaluate the safety, pharmacokinetics and pharmacodynamics of DS-1040 in healthy subjects. Subjects/Methods Young (18-45 years) or elderly (65-75 years) subjects (N = 103) were randomized to receive single ascending doses of DS-1040 ranging from 0.1 mg to 40 mg, or placebo, administered either as a 0.5-h intravenous infusion or as a 24-h continuous infusion. Results All doses of DS-1040 were tolerated, and no serious adverse events (AEs) or discontinuations resulting from AEs occurred during the study. Bleeding time remained within the normal range for all doses tested in all subjects. Plasma exposure of DS-1040 increased proportionally with increase in dose. Elderly subjects had higher exposures to DS-1040 and prolonged elimination times, probably because of decreased renal clearance. DS-1040 caused a substantial dose-dependent and time-dependent decrease in TAFIa activity and in 50% clot lysis time. The levels of D-dimer, indicative of endogenous fibrinolysis, increased in some individuals following DS-1040 treatment. No effects of DS-1040 on coagulation parameters or platelet aggregation were observed. Conclusions The novel fibrinolysis-enhancing agent DS-1040 has favorable pharmacokinetic/pharmacodynamic properties and a favorable safety profile, warranting further clinical development.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis/drug effects , Fibrinolytic Agents/administration & dosage , Protease Inhibitors/administration & dosage , Adolescent , Adult , Age Factors , Aged , Blood Coagulation Tests , Carboxypeptidase B2/metabolism , Dose-Response Relationship, Drug , Female , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/pharmacokinetics , Healthy Volunteers , Hemorrhage/chemically induced , Humans , Infusions, Intravenous , Male , Middle Aged , Protease Inhibitors/adverse effects , Protease Inhibitors/pharmacokinetics , Risk Factors , Young Adult
11.
Clin Exp Metastasis ; 34(2): 155-169, 2017 02.
Article in English | MEDLINE | ID: mdl-28124276

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI) is a basic carboxypeptidase zymogen present in blood plasma. Proteolytic activation of TAFI by thrombin, thrombin in complex with the endothelial cell cofactor thrombomodulin, or plasmin results in an enzyme (TAFIa) that removes carboxyl-terminal lysine residues from protein and peptide substrates, including cell-surface plasminogen receptors. TAFIa is therefore capable of inhibiting plasminogen activation in the pericellular milieu. Since plasminogen activation has been linked to angiogenesis, TAFIa could therefore have anti-angiogenic properties, and indeed TAFIa has been shown to inhibit endothelial tube formation in a fibrin matrix. In this study, the TAFI pathway was manipulated by providing exogenous TAFI or TAFIa or by adding a potent and specific inhibitor of TAFIa. We found that TAFIa elicited a series of anti-angiogenic responses by endothelial cells, including decreased endothelial cell proliferation, cell invasion, cell migration, tube formation, and collagen degradation. Moreover, TAFIa decreased tube formation and proteolysis in endothelial cell culture grown alone and in co-culture with breast cancer cell lines. In accordance with these findings, inhibition of TAFIa increased secretion of matrix metalloprotease proenzymes by endothelial and breast cancer cells. Finally, treatment of endothelial cells with TAFIa significantly inhibited plasminogen activation. Taken together our results suggest a novel role for TAFI in inhibiting tumour angiogenic behaviors in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Carboxypeptidase B2/physiology , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Carboxypeptidase B2/antagonists & inhibitors , Carboxypeptidase B2/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Collagen Type IV/metabolism , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Enzyme Precursors/pharmacology , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Plasminogen/antagonists & inhibitors , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
12.
J Med Chem ; 59(20): 9567-9573, 2016 Oct 27.
Article in English | MEDLINE | ID: mdl-27749053

ABSTRACT

Previously disclosed TAFIa inhibitors having a urea zinc-binding motif were used as the starting point for the development of a novel class of highly potent inhibitors having a sulfamide zinc-binding motif. High-resolution X-ray cocrystal structures were used to optimize the structures and reveal a highly unusual sulfamide configuration. A selected sulfamide was profiled in vitro and in vivo and displayed a promising ADMET profile.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Zinc/chemistry , Animals , Carboxypeptidase B2/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Mice , Microsomes/chemistry , Microsomes/metabolism , Models, Molecular , Molecular Structure , Protease Inhibitors/chemical synthesis , Rats , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , Sulfonamides/chemistry
13.
Sci Rep ; 6: 32958, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27604544

ABSTRACT

Mature thrombin activatable fibrinolysis inhibitor (TAFIa) is a carboxypeptidase that stabilizes fibrin clots by removing C-terminal arginines and lysines from partially degraded fibrin. Inhibition of TAFIa stimulates the degradation of fibrin clots and may help to prevent thrombosis. Applying a lead finding approach based on literature-mining, we discovered that anabaenopeptins, cyclic peptides produced by cyanobacteria, were potent inhibitors of TAFIa with IC50 values as low as 1.5 nM. We describe the isolation and structure elucidation of 20 anabaenopeptins, including 13 novel congeners, as well as their pronounced structure-activity relationships (SAR) with respect to inhibition of TAFIa. Crystal structures of the anabaenopeptins B, C and F bound to the surrogate protease carboxypeptidase B revealed the binding modes of these large (~850 Da) compounds in detail and explained the observed SAR, i.e. the strong dependence of the potency on a basic (Arg, Lys) exocyclic residue that addressed the S1' binding pocket, and a broad tolerance towards substitutions in the pentacyclic ring that acted as a plug of the active site.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis/drug effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/pharmacology , Carboxypeptidase B/antagonists & inhibitors , Carboxypeptidase B/chemistry , Carboxypeptidase B2/chemistry , Catalytic Domain , Crystallization , Crystallography, X-Ray , Cyanobacteria/chemistry , Humans , Models, Molecular , Peptides, Cyclic/isolation & purification , Structure-Activity Relationship
14.
Arterioscler Thromb Vasc Biol ; 36(6): 1293-301, 2016 06.
Article in English | MEDLINE | ID: mdl-27102961

ABSTRACT

OBJECTIVE: The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) remains to be elucidated. Thrombin-activatable fibrinolysis inhibitor (TAFI) inhibits fibrinolysis. It remains to be elucidated whether TAFI is directly involved in the pathogenesis of CTEPH. We examined potential involvement of TAFI in the pathogenesis of CTEPH in humans. APPROACH AND RESULTS: We enrolled 68 consecutive patients undergoing right heart catheterization in our hospital, including those with CTEPH (n=27), those with pulmonary arterial hypertension (n=22), and controls (non-pulmonary hypertension, n=19). Whole blood clot lysis assay showed that the extent of clot remaining after 4 hours was significantly higher in CTEPH compared with pulmonary arterial hypertension or controls (41.9 versus 26.5 and 24.6%, both P<0.01). Moreover, plasma levels of TAFI were significantly higher in CTEPH than in pulmonary arterial hypertension or controls (19.4±4.2 versus 16.1±4.5 or 16.3±3.3 µg/mL, both P<0.05), which remained unchanged even after hemodynamic improvement by percutaneous transluminal pulmonary angioplasty. Furthermore, the extent of clot remaining after 4 hours was significantly improved with CPI-2KR (an inhibitor of activated TAFI) or prostaglandin E1 (an inhibitor of activation of platelets). Importantly, plasma levels of TAFI were significantly correlated with the extent of clot remaining after 4 hours. In addition, the extent of clot remaining after 4 hours was improved with an activated TAFI inhibitor. CONCLUSIONS: These results indicate that plasma levels of TAFI are elevated in patients with CTEPH and are correlated with resistance to clot lysis in those patients.


Subject(s)
Blood Platelets/enzymology , Carboxypeptidase B2/blood , Fibrinolysis , Hypertension, Pulmonary/blood , Pulmonary Embolism/blood , Adult , Aged , Biomarkers/blood , Blood Coagulation Tests , Blood Platelets/drug effects , Carboxypeptidase B2/antagonists & inhibitors , Carboxypeptidase B2/genetics , Cardiac Catheterization , Case-Control Studies , Chronic Disease , Female , Fibrinolysis/drug effects , Gene Frequency , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Male , Middle Aged , Polymorphism, Single Nucleotide , Protease Inhibitors/pharmacology , Pulmonary Embolism/complications , Pulmonary Embolism/diagnosis , Pulmonary Embolism/enzymology , Time Factors , Up-Regulation
15.
J Thromb Haemost ; 13(11): 2093-101, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26341360

ABSTRACT

BACKGROUND: Thrombin-activatable fibrinolysis inhibitor (TAFI) is a risk factor for coronary heart disease. TAFI is proteolytically activated by thrombin, the thrombin-thrombomodulin complex and plasmin. Once active, it dampens fibrinolysis and inflammation. The aim of this study was to generate TAFI-derived peptides that specifically modulate TAFI activation and activity. METHODS: Thirty-four overlapping TAFI peptides, and modifications thereof, were synthesized. The effects of these peptides on TAFI activation and TAFIa activity were determined. In addition, the binding of the peptides to thrombin were determined. RESULTS: Four peptides (peptides 2, 18, 19 and 34) inhibited TAFI activation and two peptides (peptides 14 and 24) inhibited TAFIa activity directly. Peptide 2 (Arg12-Glu28) and peptide 34 (Cys383-Val401) inhibited TAFI activation by the thrombin-thrombomodulin complex with IC50 values of 7.3 ± 1.8 and 6.1 ± 0.9 µm, respectively. However, no inhibition was observed in the absence of thrombomodulin. This suggests that the regions Arg12-Glu28 and Cys383-Val401 in TAFI are involved in thrombomodulin-mediated TAFI activation. Peptide 18 (Gly205-Ser221) and peptide 19 (Arg214-Asp232) inhibited TAFI activation by thrombin and the thrombin-thrombomodulin complex. Furthermore, these peptides bound to thrombin (KD : 1.5 ± 0.4 and 0.52 ± 0.07 µm for peptides 18 and 19, respectively), suggesting that Gly205-Asp232 of TAFI is involved in binding to thrombin. Peptide 14 (His159-His175) inhibited TAFIa activity. The inhibition was TAFIa specific, because no effect on the homologous enzyme carboxypeptidase B was observed. CONCLUSIONS: Thrombin-activatable fibrinolysis inhibitor-derived peptides show promise as new tools to modulate TAFI activation and TAFIa activity. Furthermore, these peptides revealed potential binding sites on TAFI for thrombin and the thrombin-thrombomodulin complex.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Peptide Fragments/pharmacology , Thrombin/pharmacology , Amino Acid Sequence , Carboxypeptidase B2/chemistry , Enzyme Activation/drug effects , Half-Life , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Protein Binding , Protein Conformation , Structure-Activity Relationship , Surface Plasmon Resonance , Thrombin/metabolism , Thrombomodulin/metabolism
16.
J Med Chem ; 58(11): 4839-44, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25990761

ABSTRACT

Anabaenopeptins isolated from cyanobacteria were identified as inhibitors of carboxypeptidase TAFIa. Cocrystal structures of these macrocyclic natural product inhibitors in a modified porcine carboxypeptidase B revealed their binding mode and provided the basis for the rational design of small molecule inhibitors with a previously unknown central urea motif. Optimization based on these design concepts allowed for a rapid evaluation of the SAR and delivered potent small molecule inhibitors of TAFIa with a promising overall profile.


Subject(s)
Biological Products/pharmacology , Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis/drug effects , Microsomes/drug effects , Peptides, Cyclic/pharmacology , Small Molecule Libraries/pharmacology , Animals , Biological Products/chemistry , Cells, Cultured , Crystallography, X-Ray , Cyanobacteria/chemistry , Humans , Mice , Models, Molecular , Molecular Structure , Peptides, Cyclic/chemistry , Rats , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Swine
17.
J Am Soc Nephrol ; 26(8): 1925-37, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25411467

ABSTRACT

Uncontrolled diabetes, inflammation, and hypertension are key contributors to progressive renal fibrosis and subsequent loss of renal function. Reduced fibrinolysis appears to be a feature of ESRD, but its contribution to the fibrotic program has not been extensively studied. Here, we show that in patients with CKD, the activity levels of serum thrombin-activated fibrinolysis inhibitor and plasmin strongly correlated with the degree of renal function impairment. We made similar observations in rats after subtotal nephrectomy and tested whether pharmacologic inhibition of thrombin-activated fibrinolysis inhibitor with UK-396082 could reduce renal fibrosis and improve renal function. Compared with untreated animals, UK-396082-treated animals had reduced glomerular and tubulointerstitial fibrosis after subtotal nephrectomy. Renal function, as measured by an increase in creatinine clearance, was maintained and the rate of increase in proteinuria was reduced in UK-396082-treated animals. Furthermore, cumulative survival improved from 16% to 80% with inhibition of thrombin-activated fibrinolysis inhibitor. Taken together, these data support the importance of the fibrinolytic axis in regulating renal fibrosis and point to a potentially important therapeutic role for suppression of thrombin-activated fibrinolysis inhibitor activity.


Subject(s)
Amino Acids/therapeutic use , Carboxypeptidase B2/blood , Fibrinolysin/metabolism , Imidazoles/therapeutic use , Renal Insufficiency, Chronic/blood , Adult , Aged , Amino Acids/pharmacology , Animals , Biomarkers/urine , Carboxypeptidase B2/antagonists & inhibitors , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Fibrosis , Humans , Imidazoles/pharmacology , Kidney/drug effects , Kidney/pathology , Kidney Function Tests , Male , Middle Aged , Nephrectomy , Random Allocation , Rats, Wistar , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/urine
18.
Cerebrovasc Dis ; 38(4): 268-75, 2014.
Article in English | MEDLINE | ID: mdl-25401979

ABSTRACT

BACKGROUND: Since thrombolysis is the only approved intervention for ischemic stroke, improving its efficacy and safety is a therapeutic aim of considerable interest. The activated form of thrombin activatable fibrinolysis inhibitor (TAFI) has antifibrinolytic effects, and inhibition of TAFI might thus favor recanalization. The present study compared efficacy between TAFI inhibition alone and TAFI inhibition in combination with rtPA at a suboptimal dose, in a murine model of thromboembolic stroke. METHODS: Focal ischemia was induced in mice by thrombin injection in the middle cerebral artery. Animals were placed within the magnet immediately after surgery for baseline MRI (H0). MRI examination comprised diffusion-weighted imaging (DWI), perfusion-weighted imaging (PWI), and T2-weighted imaging (T2-WI). Animals were randomly assigned to 1 of 5 treatment groups: saline, rtPA 5 mg/kg (tPA(5): suboptimal or low dose), rtPA 10 mg/kg (tPA(10): standard dose), TAFI-I 100 mg/kg (TAFI-I), and rtPA 5 mg/kg + TAFI-I 100 mg/kg (tPA(5) + TAFI-I). Treatments were administered inside the magnet, via a catheter placed in the tail vein, using a power injector, as 10% bolus and 90% infusion over a period of 20 min. MRI examination was repeated at 3 h (H3) and 24 h (H24) after surgery. Therapeutic benefit was evaluated by: (1) improvement of reperfusion and (2) reduction in final lesion size. Microhemorrhages were assessed as black spots on T2-WI at H24. Animals were sacrificed after the last MR examination. The surgeon and all investigators were blinded to treatment allocation. RESULTS: A total of 104 mice were operated on. Forty four of these were excluded from the study and 27 from the analysis, according to a priori defined criteria (no lesion or no mismatch), leading to the following distribution: saline (n = 6), tPA(5) (n = 8), tPA(10) (n = 7), TAFI-I (n = 7), and TAFI-I + tPA(5) (n = 5). Standard-dose rtPA treatment (tPA(10)) significantly improved lesion regression between H0 and H24 compared to saline (-57 ± 18% vs. -36 ± 21%, p = 0.03), which treatment with rtPA(5) or TAFI-I alone did not. On the other hand, combined treatment with tPA(5) + TAFI-I showed only a trend toward lesion regression (-49 ± 26%), similarly to treatment with tPA(10), but not significantly different from saline (p = 0.46). Nine animals showed microhemorrhage on T2-WI at H24. These animals were evenly distributed between groups. CONCLUSIONS: The present study showed that the combination of TAFI-I with a suboptimal dose of rtPA is not as effective as the standard dose of rtPA, while TAFI inhibition alone is not effective at all. The thromboembolic model is of particular interest in assessing rtPA association to improve thrombolysis, especially when coupled with longitudinal MRI assessment.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Fibrinolytic Agents/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Thromboembolism/drug therapy , Thrombolytic Therapy , Tissue Plasminogen Activator/administration & dosage , Animals , Carboxypeptidase B2/blood , Cerebrovascular Circulation/drug effects , Diffusion Magnetic Resonance Imaging , Disease Models, Animal , Drug Therapy, Combination , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/chemically induced , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Perfusion Imaging/methods , Recombinant Proteins/administration & dosage , Thrombin , Thromboembolism/blood , Thromboembolism/chemically induced , Thromboembolism/pathology , Thromboembolism/physiopathology
19.
Bioorg Med Chem ; 22(7): 2261-8, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24588961

ABSTRACT

A series of 4,5,6,7-tetrahydro-1H-benzimidazole-5-carboxylic acid and 5,6,7,8-tetrahydroimidazo[1,2-a]pyridine-7-carboxylic acid derivatives designed as inhibitors of TAFIa has been prepared via a common hydrogenation-alkylation sequence starting from the appropriate benzimidazole and imidazopyridine system. We present a successful design strategy using a conformational restriction approach resulting in potent and selective inhibitors of TAFIa. The X-ray structure of compound 5 in complex with a H333Y/H335Q double mutant TAFI indicate that the conformational restriction is responsible for the observed potency increase.


Subject(s)
Benzimidazoles/pharmacology , Carboxypeptidase B2/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Caco-2 Cells , Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Conformation , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship
20.
J Thromb Haemost ; 12(2): 229-36, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24354544

ABSTRACT

BACKGROUND: Down-regulation of fibrinolysis due to cleavage of C-terminal lysine residues from partially degraded fibrin is mainly exerted by the carboxypeptidase activity of activated thrombin-activatable fibrinolysis inhibitor (TAFIa). Recently, some intrinsic carboxypeptidase activity (i.e. zymogen activity) was reported for the proenzyme (TAFI); however, there is some discussion about its ability to cleave high molecular weight substrates. OBJECTIVE: We aimed to identify and characterize nanobodies toward mouse TAFI (mTAFI) that stimulate the zymogen activity and to test their effect in an in vitro clot lysis assay and an in vivo mouse thromboembolism model. METHODS AND RESULTS: Screening of a library of nanobodies toward mTAFI revealed one nanobody (VHH-mTAFI-i49) that significantly stimulates the zymogen activity of mTAFI from undetectable (< 0.35 U mg⁻¹) to 4.4 U mg⁻¹ (at a 16-fold molar ratio over mTAFI). The generated carboxypeptidase activity is unstable at 37 °C. Incubation of mTAFI with VHH-mTAFI-i49 revealed a time-dependent reduced activatability of mTAFI. Epitope mapping revealed that Arg227 and Lys212 are important for the nanobody/mTAFI interaction and suggest destabilization of mTAFI by disrupting the stabilizing interaction between the activation peptide and the dynamic flap region. In vitro clot lysis experiments revealed an enhanced clot lysis due to a reduced activation of mTAFI during clot formation. In vivo application of VHH-mTAFI-i49 in a mouse thromboembolism model decreased dose-dependently the fibrin deposition in the lungs of thromboembolism-induced mice. CONCLUSION: The novel, nanobody-induced, reduced activatability of mTAFI demonstrates to be a very potent approach to enhance clot lysis.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Single-Domain Antibodies/physiology , Animals , Mice
SELECTION OF CITATIONS
SEARCH DETAIL