Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Molecules ; 27(4)2022 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-35209023

RESUMEN

In the modern world, complications caused by disorders in the blood coagulation system are found in almost all areas of medicine. Thus, the development of new, more advanced drugs that can prevent pathological conditions without disrupting normal hemostasis is an urgent task. The blood coagulation factor XIIa is one of the most promising therapeutic targets for the development of anticoagulants based on its inhibitors. The initial stage of drug development is directly related to computational methods of searching for a lead compound. In this study, docking followed by quantum chemical calculations was used to search for noncovalent low-molecular-weight factor XIIa inhibitors in a focused library of druglike compounds. As a result of the study, four low-molecular-weight compounds were experimentally confirmed as factor XIIa inhibitors. Selectivity testing revealed that two of the identified factor XIIa inhibitors were selective over the coagulation factors Xa and XIa.


Asunto(s)
Proteínas Sanguíneas , Factor XIIa , Simulación del Acoplamiento Molecular , Proteínas Sanguíneas/síntesis química , Proteínas Sanguíneas/química , Factor XIIa/antagonistas & inhibidores , Factor XIIa/química , Humanos
2.
Thromb Haemost ; 122(2): 196-207, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34619795

RESUMEN

BACKGROUND: 3F7 is a monoclonal antibody targeting the enzymatic pocket of activated factor XII (FXIIa), thereby inhibiting its catalytic activity. Given the emerging role of FXIIa in promoting thromboinflammation, along with its apparent redundancy for hemostasis, the selective inhibition of FXIIa represents a novel and highly attractive approach targeting pathogenic processes that cause thromboinflammation-driven cardiovascular diseases. METHODS: The effects of FXIIa inhibition were investigated using three distinct mouse models of cardiovascular disease-angiotensin II-induced abdominal aortic aneurysm (AAA), an ApoE-/- model of atherosclerosis, and a tandem stenosis model of atherosclerotic plaque instability. 3F7 or its isotype control, BM4, was administered to mice (10 mg/kg) on alternate days for 4 to 8 weeks, depending on the experimental model. Mice were examined for the development and size of AAAs, or the burden and instability of atherosclerosis and associated markers of inflammation. RESULTS: Inhibition of FXIIa resulted in a reduced incidence of larger AAAs, with less acute aortic ruptures and an associated fibro-protective phenotype. FXIIa inhibition also decreased stable atherosclerotic plaque burden and achieved plaque stabilization associated with increased deposition of fibrous structures, a >2-fold thicker fibrous cap, increased cap-to-core ratio, and reduction in localized and systemic inflammatory markers. CONCLUSION: Inhibition of FXIIa attenuates disease severity across three mouse models of thromboinflammation-driven cardiovascular diseases. Specifically, the FXIIa-inhibiting monoclonal antibody 3F7 reduces AAA severity, inhibits the development of atherosclerosis, and stabilizes vulnerable plaques. Ultimately, clinical trials in patients with cardiovascular diseases such as AAA and atherosclerosis are warranted to demonstrate the therapeutic potential of FXIIa inhibition.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Aneurisma de la Aorta Abdominal/prevención & control , Aterosclerosis/prevención & control , Factor XIIa/antagonistas & inhibidores , Placa Aterosclerótica/metabolismo , Animales , Aneurisma de la Aorta Abdominal/epidemiología , Apolipoproteínas E , Modelos Animales de Enfermedad , Inflamación , Masculino , Ratones
3.
ChemMedChem ; 16(24): 3672-3690, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34278727

RESUMEN

Herein we report a microscale parallel synthetic approach allowing for rapid access to libraries of N-acylated aminotriazoles and screening of their inhibitory activity against factor XIIa (FXIIa) and thrombin, which are targets for antithrombotic drugs. This approach, in combination with post-screening structure optimization, yielded a potent 7 nM inhibitor of FXIIa and a 25 nM thrombin inhibitor; both compounds showed no inhibition of the other tested serine proteases. Selected N-acylated aminotriazoles exhibited anticoagulant properties in vitro influencing the intrinsic blood coagulation pathway, but not extrinsic coagulation. Mechanistic studies of FXIIa inhibition suggested that synthesized N-acylated aminotriazoles are covalent inhibitors of FXIIa. These synthesized compounds may serve as a promising starting point for the development of novel antithrombotic drugs.


Asunto(s)
Amitrol (Herbicida)/farmacología , Anticoagulantes/farmacología , Factor XIIa/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/farmacología , Trombina/antagonistas & inhibidores , Acilación , Amitrol (Herbicida)/síntesis química , Amitrol (Herbicida)/química , Anticoagulantes/síntesis química , Anticoagulantes/química , Coagulación Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Factor XIIa/metabolismo , Humanos , Estructura Molecular , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/química , Relación Estructura-Actividad , Trombina/metabolismo
4.
Expert Opin Ther Pat ; 31(12): 1155-1176, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34142629

RESUMEN

Introduction: Blood coagulation factor XII (FXII) is an emerging and potentially safe drug target, which dysregulation is associated with thrombosis, hereditary angioedema, and (neuro)inflammation. At the same time, FXII-deficiency is practically asymptomatic. Industrial and academic institutions have developed a number of potential therapeutic agents targeting either FXII zymogen or its active form FXIIa for the treatment of thrombotic and inflammatory conditions associated with the activity of this enzyme.Areas covered: A short overview of the FXII(a) structure and function, underlining its suitability as a drug target, is given. The article reviews patents reported over the last three decades on FXII(a)-targeting therapeutic agents. These agents include small molecules, proteins, peptides, oligonucleotides, siRNAs, and monoclonal antibodies.Expert opinion: The performed analysis of patents revealed that many FXII(a) inhibitors are in the early preclinical stage, while several already showed efficacy in vivo animal models of thrombosis, sepsis, hereditary angioedema, and multiple sclerosis. Two anti-FXIIa agents namely tick protein Ir-CPI and monoclonal antibody CSL312 are currently in human clinical trials. The results of these trials and further studies of FXII(a) pathophysiological functions will encourage the development of new FXII(a) inhibitors.


Asunto(s)
Factor XII/antagonistas & inhibidores , Factor XIIa/antagonistas & inhibidores , Angioedemas Hereditarios/tratamiento farmacológico , Angioedemas Hereditarios/patología , Animales , Desarrollo de Medicamentos , Factor XII/metabolismo , Factor XIIa/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Patentes como Asunto , Trombosis/tratamiento farmacológico , Trombosis/patología
5.
J Med Chem ; 64(11): 7853-7876, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34044534

RESUMEN

The contact system comprises a series of serine proteases that mediate procoagulant and proinflammatory activities via the intrinsic pathway of coagulation and the kallikrein-kinin system, respectively. Inhibition of Factor XIIa (FXIIa), an initiator of the contact system, has been demonstrated to lead to thrombo-protection and anti-inflammatory effects in animal models and serves as a potentially safer target for the development of antithrombotics. Herein, we describe the use of the Randomised Nonstandard Peptide Integrated Discovery (RaPID) mRNA display technology to identify a series of potent and selective cyclic peptide inhibitors of FXIIa. Cyclic peptides were evaluated in vitro, and three lead compounds exhibited significant prolongation of aPTT, a reduction in thrombin generation, and an inhibition of bradykinin formation. We also describe our efforts to identify the critical residues for binding FXIIa through alanine scanning, analogue generation, and via in silico methods to predict the binding mode of our lead cyclic peptide inhibitors.


Asunto(s)
Factor XIIa/antagonistas & inhibidores , Péptidos Cíclicos/química , ARN Mensajero/metabolismo , Inhibidores de Serina Proteinasa/química , Sitios de Unión , Factor XIIa/metabolismo , Biblioteca de Genes , Código Genético , Humanos , Concentración 50 Inhibidora , Calicreínas/química , Calicreínas/metabolismo , Simulación de Dinámica Molecular , Tiempo de Tromboplastina Parcial , Péptidos Cíclicos/metabolismo , Estabilidad Proteica , Tiempo de Protrombina , Puromicina/química , ARN Mensajero/química , Inhibidores de Serina Proteinasa/metabolismo , Relación Estructura-Actividad
6.
J Thromb Haemost ; 19(2): 323-329, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33047454

RESUMEN

Clinical practice shows that a critical unmet need in the field of medical device-associated thrombosis prevention is the availability of an anticoagulant therapy without hemorrhagic risk. In the quest for new drugs that are at least as effective as those currently available, while avoiding bleeding complications, molecules that target nearly every step of the coagulation pathway have been developed. Among these molecules, inhibitors of factor XII (FXII) or factor XI (FXI) are promising alternatives as deficiencies in these factors protect against thrombosis without causing spontaneous hemorrhage, as revealed by epidemiological and preclinical data. Ixodes ricinus-contact phase inhibitor (Ir-CPI), a new anticoagulant candidate with an innovative mechanism of action could be this ideal anticoagulant agent for safe prevention from clotting on medical devices. This protein, which selectively binds to FXIIa, FXIa, and plasma kallikrein and inhibits the reciprocal activation of FXII, prekallikrein, and FXI in human plasma, was shown to prevent thrombosis in an ovine cardiopulmonary bypass system associated with cardiac surgeries. Furthermore, as opposed to unfractionated heparin, Ir-CPI appears to be devoid of bleeding risk. This review outlines the rationale for targeting upstream coagulation factors in order to prevent medical device-associated thrombosis; examines the novel approaches under development; and focuses on Ir-CPI, which shows promising properties in the field of thrombosis prevention.


Asunto(s)
Factor XIIa , Factor XIa , Trombosis/prevención & control , Animales , Coagulación Sanguínea , Factor XI , Factor XII , Factor XIIa/antagonistas & inhibidores , Factor XIa/antagonistas & inhibidores , Heparina , Humanos , Ovinos
7.
J Med Chem ; 63(21): 13159-13186, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33089691

RESUMEN

We herein report the conventional and microscale parallel synthesis of selective inhibitors of human blood coagulation factor XIIa and thrombin exhibiting a 1,2,4-triazol-5-amine scaffold. Structural variations of this scaffold allowed identifying derivative 21i, a potent 29 nM inhibitor of FXIIa, with improved selectivity over other tested serine proteases and also finding compound 21m with 27 nM inhibitory activity toward thrombin. For the first time, acylated 1,2,4-triazol-5-amines were proved to have anticoagulant properties and the ability to affect thrombin- and cancer-cell-induced platelet aggregation. Performed mass spectrometric analysis and molecular modeling allowed us to discover previously unknown interactions between the synthesized inhibitors and the active site of FXIIa, which uncovered the mechanistic details of FXIIa inhibition. Synthesized compounds represent a promising starting point for the development of novel antithrombotic drugs or chemical tools for studying the role of FXIIa and thrombin in physiological and pathological processes.


Asunto(s)
Aminas/química , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor XIIa/metabolismo , Trombina/metabolismo , Aminas/síntesis química , Aminas/metabolismo , Anticoagulantes/síntesis química , Anticoagulantes/metabolismo , Sitios de Unión , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Factor XIIa/antagonistas & inhibidores , Humanos , Concentración 50 Inhibidora , Simulación de Dinámica Molecular , Agregación Plaquetaria/efectos de los fármacos , Relación Estructura-Actividad , Trombina/antagonistas & inhibidores , Triazoles/química
8.
Eur J Med Chem ; 208: 112753, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-32883641

RESUMEN

Coagulation factor XII (FXII), a S1A serine protease, was discovered more than fifty years ago. However, its in vivo functions and its three-dimensional structure started to be disclosed in the last decade. FXII was found at the crosstalk of several physiological pathways including the intrinsic coagulation pathway, the kallikrein-kinin system, and the immune response. The FXII inhibition emerges as a therapeutic strategy for the safe prevention of artificial surface-induced thrombosis and in patients suffering from hereditary angioedema. The anti-FXII antibody garadacimab discovered by phage-display library technology is actually under phase II clinical evaluation for the prophylactic treatment of hereditary angioedema. The implication of FXII in neuro-inflammatory and neurodegenerative disorders is also an emerging research field. The FXII or FXIIa inhibitors currently under development include peptides, proteins, antibodies, RNA-based technologies, and, to a lesser extent, small-molecular weight inhibitors. Most of them are proteins, mainly isolated from hematophagous arthropods and plants. The discovery and development of these FXII inhibitors and their potential indications are discussed in the review.


Asunto(s)
Anticoagulantes/farmacología , Factor XII/antagonistas & inhibidores , Factor XIIa/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/farmacología , Animales , Anticoagulantes/química , Descubrimiento de Drogas , Factor XII/química , Factor XIIa/química , Humanos , Inhibidores de Serina Proteinasa/química
9.
Nat Commun ; 11(1): 3890, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32753636

RESUMEN

Inhibiting thrombosis without generating bleeding risks is a major challenge in medicine. A promising solution may be the inhibition of coagulation factor XII (FXII), because its knock-out or inhibition in animals reduced thrombosis without causing abnormal bleeding. Herein, we have engineered a macrocyclic peptide inhibitor of activated FXII (FXIIa) with sub-nanomolar activity (Ki = 370 ± 40 pM) and a high stability (t1/2 > 5 days in plasma), allowing for the preclinical evaluation of a first synthetic FXIIa inhibitor. This 1899 Da molecule, termed FXII900, efficiently blocks FXIIa in mice, rabbits, and pigs. We found that it reduces ferric-chloride-induced experimental thrombosis in mice and suppresses blood coagulation in an extracorporeal membrane oxygenation (ECMO) setting in rabbits, all without increasing the bleeding risk. This shows that FXIIa activity is controllable in vivo with a synthetic inhibitor, and that the inhibitor FXII900 is a promising candidate for safe thromboprotection in acute medical conditions.


Asunto(s)
Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor XIIa/antagonistas & inhibidores , Péptidos Cíclicos/efectos de los fármacos , Trombosis/prevención & control , Animales , Cloruros/efectos adversos , Clonación Molecular , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Oxigenación por Membrana Extracorpórea/métodos , Factor XII/antagonistas & inhibidores , Femenino , Compuestos Férricos/efectos adversos , Humanos , Pulmón , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Conejos , Proteínas Recombinantes/farmacología , Porcinos
10.
Comput Math Methods Med ; 2020: 2852051, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32549905

RESUMEN

Human coagulation factor XIIa (FXIIa) is a trypsin-like serine protease that is involved in pathologic thrombosis. As a potential target for designing safe anticoagulants, FXIIa has received a great deal of interest in recent years. In the present study, we employed virtual high-throughput screening of 500,064 compounds within Enamine database to acquire the most potential inhibitors of FXIIa. Subsequently, 18 compounds with significant binding energy (from -65.195 to -15.726 kcal/mol) were selected, and their ADMET properties were predicted to select representative inhibitors. Three compounds (Z1225120358, Z432246974, and Z146790068) exhibited excellent binding affinity and druggability. MD simulation for FXIIa-ligand complexes was carried out to reveal the stability and inhibition mechanism of these three compounds. Through the inhibition of activated factor XIIa assay, we tested the activity of five compounds Z1225120358, Z432246974, Z45287215, Z30974175, and Z146790068, with pIC50 values of 9.3∗10-7, 3.0∗10-5, 7.8∗10-7, 8.7∗10-7, and 1.3∗10-6 M, respectively; the AMDET properties of Z45287215 and Z30974175 show not well but have better inhibition activity. We also found that compounds Z1225120358, Z45287215, Z30974175, and Z146790068 could be more inhibition of FXIIa than Z432246974. Collectively, compounds Z1225120358, Z45287215, Z30974175, and Z146790068 were anticipated to be promising drug candidates for inhibition of FXIIa.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Factor XIIa/antagonistas & inhibidores , Factor XIIa/química , Sitios de Unión , Biología Computacional , Bases de Datos Farmacéuticas , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/estadística & datos numéricos , Factor XIIa/metabolismo , Ensayos Analíticos de Alto Rendimiento/estadística & datos numéricos , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Interfaz Usuario-Computador
11.
J Am Coll Cardiol ; 74(17): 2178-2189, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31648711

RESUMEN

BACKGROUND: Exposure of blood to polyanionic artificial surfaces, for example, during cardiopulmonary bypass (CPB), induces a highly procoagulant condition requiring strong anticoagulation. Unfractionated heparin (UFH) is currently used during CPB but can lead to serious bleeding complications or development of a hypercoagulable state culminating in life-threatening thrombosis, highlighting the need for safer antithrombotics. Ixodes ricinus contact phase inhibitor (Ir-CPI) is a protein expressed by I. ricinus ticks, which specifically inhibits both factors XIIa and XIa, 2 factors contributing to thrombotic disease while playing a limited role in hemostasis. OBJECTIVES: This study assessed the antithrombotic activity of Ir-CPI in animal contact phase-initiated thrombosis models, including CPB. The safety of Ir-CPI also was evaluated. METHODS: The authors evaluated the antithrombotic activity of Ir-CPI by using in vitro catheter-induced clotting assays and rabbit experimental models of catheter occlusion and arteriovenous shunt. During CPB with cardiac surgery in sheep, the clinical applicability of Ir-CPI was investigated and its efficacy compared to that of UFH using an uncoated system suitable for adult therapy. Taking advantage of the similar hemostatic properties of pigs and humans, the authors performed pig liver bleeding assays to evaluate the safety of Ir-CPI. RESULTS: Ir-CPI prevented clotting in catheter and arteriovenous shunt rabbit models. During CPB, Ir-CPI was as efficient as UFH in preventing clot formation within the extracorporeal circuit and maintained physiological parameters during and post-surgery. Unlike UFH, Ir-CPI did not promote bleeding. CONCLUSIONS: Preclinical animal models used in this study showed that Ir-CPI is an effective and safe antithrombotic agent that provides a clinically relevant approach to thrombosis prevention in bypass systems, including highly thrombogenic CPB.


Asunto(s)
Anticoagulantes/uso terapéutico , Puente Cardiopulmonar/métodos , Factor XIIa/antagonistas & inhibidores , Factor XIa/antagonistas & inhibidores , Animales , Coagulación Sanguínea , Proteínas Sanguíneas/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Fibrinolíticos , Hemorragia/tratamiento farmacológico , Hemostasis , Heparina/uso terapéutico , Humanos , Ixodes , Conejos , Ovinos , Porcinos , Trombosis/prevención & control , Garrapatas
12.
Chem Biol Drug Des ; 94(5): 1905-1918, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31148409

RESUMEN

The experimental inhibitory potency of benzamidine (BEN) paved way for further design and development of inhibitors that target ß-FXIIa. Structural dynamics of the loops and catalytic residues that encompass the binding pocket of ß-FXIIa and all serine proteases are crucial to their overall activity. Employing molecular dynamics and post-MD analysis, this study sorts to unravel the structural and molecular events that accompany the inhibitory activity of BEN on human ß-FXIIa upon selective non-covalent binding. Analysis of conformational dynamics of crucial loops revealed prominent alterations of the original conformational posture of FXIIa, evidenced by increased flexibility, decreased compactness, and an increased exposure to solvent upon binding of BEN, which could have in turn interfered with the essential roles of these loops in enhancing their procoagulation interactions with biological substrates and cofactors, altogether resulting in the consequential inactivation of FXIIa. A sustained interaction of the catalytic triad residues and key residues of the autolysis loop impeded their roles in catalysis which equally enhanced the inhibitory potency of BEN toward ß-FXIIa evidenced by a favorable binding. Findings provide essential structural and molecular insights that could facilitate the structure-based design of novel antithrombotic compounds with enhanced inhibitory activities and low therapeutic risk.


Asunto(s)
Benzamidinas/química , Coagulación Sanguínea/efectos de los fármacos , Factor XIIa/antagonistas & inhibidores , Fibrinolíticos/química , Secuencia de Aminoácidos , Dominio Catalítico , Diseño de Fármacos , Factor XIIa/química , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Termodinámica
13.
J Mol Graph Model ; 89: 225-233, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30921556

RESUMEN

Thrombosis represents a major cause of morbidity and mortality around the world. Peptides isolated from natural sources have been proven to have anticoagulant and antithrombotic properties. VITPOR AI, a 16-mer peptide, isolated from Porphyra yezoensis was reported to have anticoagulant property. In this study, the coagulation factor XIIa activity in the presence of VITPOR AI was determined. Molecular modelling was performed to investigate the interaction between peptide and FXIIa. The structure of the peptide was predicted using PEP-FOLD3 server and simulated by molecular dynamics (MD) using GROMACS package. Molecular docking was carried out using peptide-protein docking software, pepATTRACT and its stability was confirmed by MD simulations. The chromogenic substrate assay revealed that the peptide inhibited the amidolytic activity of FXIIa with IC50 of 70.24 µM. The docking result showed peptide interactions through hydrogen bonds with Pro 96, Tyr 99, Glu 146, Gly 193 and Ser 195 of FXIIa. The MD simulation demonstrated that the peptide's binding with the FXIIa was stable as it did not move away from its binding region throughout the simulation period of 100 ns Moreover, MM/PBSA analysis also indicated a stable binding between the protein and peptide. These results suggest that the inhibition of the FXIIa activity might be due to binding of the peptide to oxyanion hole of the catalytic site. Thus, VITPOR AI could be explored as a potent anticoagulant which inhibits only intrinsic pathway of coagulation cascade but does not affect the extrinsic pathway.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Factor XIIa/antagonistas & inhibidores , Factor XIIa/química , Péptidos/química , Péptidos/farmacología , Porphyra/química , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Conformación Molecular , Extractos Vegetales , Relación Estructura-Actividad
14.
Toxins (Basel) ; 10(11)2018 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-30373118

RESUMEN

Antistasin, first identified as a potent inhibitor of the blood coagulation factor Xa, is a novel family of serine protease inhibitors. In this study, we purified a novel antistasin-type inhibitor from leech Poecilobdella manillensis called poecistasin. Amino acid sequencing of this 48-amino-acid protein revealed that poecistasin was an antistasin-type inhibitor known to consist of only one domain. Poecistasin inhibited factor XIIa, kallikrein, trypsin, and elastase, but had no inhibitory effect on factor Xa and thrombin. Poecistasin showed anticoagulant activities. It prolonged the activated partial thromboplastin time and inhibited FeCl3-induced carotid artery thrombus formation, implying its potent function in helping Poecilobdella manillensis to take a blood meal from the host by inhibiting coagulation. Poecistasin also suppressed ischemic stroke symptoms in transient middle cerebral artery occlusion mice model. Our results suggest that poecistasin from the leech Poecilobdella manillensis plays a crucial role in blood-sucking and may be an excellent candidate for the development of clinical anti-thrombosis and anti-ischemic stroke medicines.


Asunto(s)
Fibrinolíticos , Sanguijuelas , Inhibidores de Serina Proteinasa , Inhibidores de Tripsina , Animales , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Factor XIIa/antagonistas & inhibidores , Factor Xa/metabolismo , Femenino , Fibrinolíticos/química , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Hormonas de Invertebrados , Ratones Endogámicos C57BL , Elastasa Pancreática/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Inhibidores de Serina Proteinasa/uso terapéutico , Trombina/metabolismo , Inhibidores de Tripsina/química , Inhibidores de Tripsina/farmacología , Inhibidores de Tripsina/uso terapéutico
15.
Chembiochem ; 19(18): 1934-1938, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-29944773

RESUMEN

A broadly applicable one-pot methodology for the facile transformation of linear peptides into tetracyclic peptides through a chemoenzymatic peptide synthesis/chemical ligation of peptides onto scaffolds/copper(I)-catalyzed reaction (CEPS/CLIPS/CuAAC; "triple-C") locking methodology is reported. Linear peptides with varying lengths (≥14 amino acids), comprising two cysteines and two azidohomoalanines (Aha), were efficiently cyclized head-to-tail by using the peptiligase variant omniligase-1 (CEPS). Subsequent ligation-cyclization with tetravalent (T41/2 ) scaffolds containing two bromomethyl groups (CLIPS) and two alkyne functionalities (CuAAC) yielded isomerically pure tetracyclic peptides. Sixteen different functional tetracycles, derived from bicyclic inhibitors against urokinase plasminogen activator (uPA) and coagulation factor XIIa (FXIIa), were successfully synthesized and their bioactivities evaluated. Two of these (FF-T41/2 ) exhibited increased inhibitory activity against FXIIa, compared with a bicyclic control peptide. The corresponding hetero-bifunctional variants (UF/FU-T41/2 ), with a single copy of each inhibitory sequence, exhibited micromolar activities against both uPA and FXIIa; thus illustrating the potential of the "bifunctional tetracyclic peptide" inhibitor concept.


Asunto(s)
Péptidos Cíclicos/síntesis química , Péptidos/química , Alanina/análogos & derivados , Alanina/química , Secuencia de Aminoácidos , Técnicas Químicas Combinatorias , Ciclización , Cisteína/química , Factor XIIa/antagonistas & inhibidores , Humanos , Modelos Moleculares , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
16.
Eur J Med Chem ; 140: 31-41, 2017 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-28918098

RESUMEN

There currently is renewed interest in blood clotting Factor XII as a potential target for thrombosis inhibition. Historically untargeted, there is little drug information with which to start drug candidate searches. Typical high-throughput screening can identify potential drug candidates, but is inefficient. Virtual high-throughput screening can be used to raise efficiency by focusing experimental efforts on compounds predicted to be active and is applied here to identify new Factor XIIa inhibitors. We combine principal component analysis, genetic algorithm and support vector machine to create the models used in the virtual high-throughput screening. In this work, experimental data from a PubChem Bioassay was used to train predictive models of Factor XIIa inhibition activity. The models created were then used to virtually screen the entire 72 million PubChem Compound database. Experimental validation of select candidates identified by this process resulted in a 42.9% hit-rate in the first-pass and 100% hit-rate in the second-pass, suggesting the effectiveness of the approach.


Asunto(s)
Algoritmos , Proteínas Sanguíneas/farmacología , Factor XIIa/antagonistas & inhibidores , Análisis de Componente Principal , Máquina de Vectores de Soporte , Proteínas Sanguíneas/síntesis química , Proteínas Sanguíneas/química , Relación Dosis-Respuesta a Droga , Factor XIIa/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
17.
J Neuroinflammation ; 14(1): 39, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28219400

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a devastating neurological condition and a frequent cause of permanent disability. Posttraumatic inflammation and brain edema formation, two pathological key events contributing to secondary brain injury, are mediated by the contact-kinin system. Activation of this pathway in the plasma is triggered by activated factor XII. Hence, we set out to study in detail the influence of activated factor XII on the abovementioned pathophysiological features of TBI. METHODS: Using a cortical cryogenic lesion model in mice, we investigated the impact of genetic deficiency of factor XII and inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused Infestin-4 on the release of bradykinin, the brain lesion size, and contact-kinin system-dependent pathological events. We determined protein levels of bradykinin, intracellular adhesion molecule-1, CC-chemokine ligand 2, and interleukin-1ß by enzyme-linked immunosorbent assays and mRNA levels of genes related to inflammation by quantitative real-time PCR. Brain lesion size was determined by tetrazolium chloride staining. Furthermore, protein levels of the tight junction protein occludin, integrity of the blood-brain barrier, and brain water content were assessed by Western blot analysis, extravasated Evans Blue dye, and the wet weight-dry weight method, respectively. Infiltration of neutrophils and microglia/activated macrophages into the injured brain lesions was quantified by immunohistological stainings. RESULTS: We show that both genetic deficiency of factor XII and inhibition of activated factor XII in mice diminish brain injury-induced bradykinin release by the contact-kinin system and minimize brain lesion size, blood-brain barrier leakage, brain edema formation, and inflammation in our brain injury model. CONCLUSIONS: Stimulation of bradykinin release by activated factor XII probably plays a prominent role in expanding secondary brain damage by promoting brain edema formation and inflammation. Pharmacological blocking of activated factor XII could be a useful therapeutic principle in the treatment of TBI-associated pathologic processes by alleviating posttraumatic inflammation and brain edema formation.


Asunto(s)
Edema Encefálico/metabolismo , Edema Encefálico/prevención & control , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/prevención & control , Factor XIIa/antagonistas & inhibidores , Factor XIIa/metabolismo , Animales , Bradiquinina/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/prevención & control , Factor XIIa/genética , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
J Pharmacol Exp Ther ; 360(3): 466-475, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28035006

RESUMEN

Coagulation Factor XII (FXII) plays a critical role in thrombosis. What is unclear is the level of enzyme occupancy of FXIIa that is needed for efficacy and the impact of FXIIa inhibition on cerebral embolism. A selective activated FXII (FXIIa) inhibitor, recombinant human albumin-tagged mutant Infestin-4 (rHA-Mut-inf), was generated to address these questions. rHA-Mut-inf displayed potency comparable to the original wild-type HA-Infestin-4 (human FXIIa inhibition constant = 0.07 and 0.12 nM, respectively), with markedly improved selectivity against Factor Xa (FXa) and plasmin. rHA-Mut-inf binds FXIIa, but not FXII zymogen, and competitively inhibits FXIIa protease activity. Its mode of action is hence akin to typical small-molecule inhibitors. Plasma shift and aPTT studies with rHA-Mut-inf demonstrated that calculated enzyme occupancy for FXIIa in achieving a putative aPTT doubling target in human, nonhuman primate, and rabbit is more than 99.0%. The effects of rHA-Mut-inf in carotid arterial thrombosis and microembolic signal (MES) in middle cerebral artery were assessed simultaneously in rabbits. Dose-dependent inhibition was observed for both arterial thrombosis and MES. The ED50 of thrombus formation was 0.17 mg/kg i.v. rHA-Mut-inf for the integrated blood flow and 0.16 mg/kg for thrombus weight; the ED50 for MES was 0.06 mg/kg. Ex vivo aPTT tracked with efficacy. In summary, our findings demonstrated that very high enzyme occupancy will be required for FXIIa active site inhibitors, highlighting the high potency and exquisite selectivity necessary for achieving efficacy in humans. Our MES studies suggest that targeting FXIIa may offer a promising strategy for stroke prevention associated with thromboembolic events.


Asunto(s)
Coagulación Sanguínea , Factor XIIa/antagonistas & inhibidores , Proteínas de Insectos/farmacología , Embolia Intracraneal , Trombosis Intracraneal , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica/farmacología , Animales , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Fibrinolíticos/farmacología , Embolia Intracraneal/sangre , Embolia Intracraneal/tratamiento farmacológico , Trombosis Intracraneal/sangre , Trombosis Intracraneal/tratamiento farmacológico , Modelos Animales , Conejos , Albúmina Sérica Humana
19.
Arterioscler Thromb Vasc Biol ; 37(1): 13-20, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27834692

RESUMEN

Coagulation factor XII (FXII, Hageman factor) is a plasma protease that in its active form (FXIIa) initiates the procoagulant and proinflammatory contact system. This name arises from FXII's unique mechanism of activation that is induced by binding (contact) to negatively charged surfaces. Various substances have the capacity to trigger FXII contact-activation in vivo including mast cell-derived heparin, misfolded protein aggregates, collagen, nucleic acids, and polyphosphate. FXII deficiency is not associated with bleeding, and for decades, the factor was considered to be dispensable for coagulation in vivo. However, despite the fact that humans and animals with deficiency in FXII have a normal hemostatic capacity, animal models revealed a critical role of FXIIa-driven coagulation in thromboembolic diseases. In addition to its role in thrombosis, FXIIa contributes to inflammation through the activation of the inflammatory bradykinin-producing kallikrein-kinin system. Pharmacological inhibition of FXII/FXIIa interferes with thrombosis and inflammation in animal models. Thus, targeting the FXIIa-driven contact system seems to be a promising and safe therapeutic anticoagulation treatment strategy, with additional anti-inflammatory effects. Here, we discuss novel functions of FXIIa in cardiovascular thrombotic and inflammatory disorders.


Asunto(s)
Antiinflamatorios/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Fibrinolíticos/uso terapéutico , Mediadores de Inflamación/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Sistema Calicreína-Quinina/efectos de los fármacos , Tromboembolia/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Factor XII/genética , Factor XII/metabolismo , Factor XIIa/antagonistas & inhibidores , Factor XIIa/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Inflamación/sangre , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/sangre , Ratones Noqueados , Terapia Molecular Dirigida , Tromboembolia/sangre , Tromboembolia/genética , Tromboembolia/inmunología
20.
PLoS One ; 11(10): e0165493, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27783665

RESUMEN

The serpin C1 inhibitor (C1-INH) is the only regulator of classical complement activation as well as the major regulator of the contact system. Its importance is demonstrated by hereditary angioedema (HAE), a severe disease with potentially life-threatening attacks due to deficiency or dysfunction of C1-INH. C1-INH replacement is the therapy of choice in HAE. In addition, C1-INH showed to have beneficial effects in other diseases characterized by inappropriate complement and contact system activation. Due to some limitations of its clinical application, there is a need for improving the efficacy of therapeutically applied C1-INH or to enhance the activity of endogenous C1-INH. Given the known potentiating effect of heparin on C1-INH, sulfated glycans (SG) may be such candidates. The aim of this study was to characterize suitable SG by evaluating structure-activity relationships. For this, more than 40 structurally distinct SG were examined for their effects on C1-INH, C1s and FXIIa. The SG turned out to potentiate the C1s inhibition by C1-INH without any direct influence on C1s. Their potentiating activity proved to depend on their degree of sulfation, molecular mass as well as glycan structure. In contrast, the SG had no effect on the FXIIa inhibition by C1-INH, but structure-dependently modulated the activity of FXIIa. Among the tested SG, ß-1,3-glucan sulfates with a Mr ≤ 10 000 were identified as most promising lead candidates for the development of a glycan-based C1-INH amplifier. In conclusion, the obtained information on structural characteristics of SG favoring C1-INH potentiation represent an useful elementary basis for the development of compounds improving the potency of C1-INH in diseases and clinical situations characterized by inappropriate activation of complement and contact system.


Asunto(s)
Proteína Inhibidora del Complemento C1/metabolismo , Factor XIIa/metabolismo , Polisacáridos/metabolismo , Alginatos/química , Alginatos/metabolismo , Activación de Complemento , Proteína Inhibidora del Complemento C1/química , Sulfato de Dextran/química , Sulfato de Dextran/metabolismo , Pruebas de Enzimas , Factor XIIa/antagonistas & inhibidores , Ácido Glucurónico/química , Ácido Glucurónico/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Ácidos Hexurónicos/química , Ácidos Hexurónicos/metabolismo , Humanos , Cinética , Polisacáridos/química , Unión Proteica , Relación Estructura-Actividad , Sulfatos/química , beta-Glucanos/química , beta-Glucanos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...