Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Molecules ; 26(14)2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34299400

RESUMEN

The goal of this study was to assess the pharmacological effects of black tea (Camellia sinensis var. assamica) water extract on human kinin-forming enzymes in vitro. Tea is a highly consumed beverage in the world. Factor XII (FXII, Hageman factor)-independent- and -dependent activation of prekallikrein to kallikrein leads to the liberation of bradykinin (BK) from high-molecular-weight kininogen (HK). The excessive BK production causes vascular endothelial and nonvascular smooth muscle cell permeability, leading to angioedema. The prevalence of angiotensin-converting enzyme inhibitor (ACEI)-induced angioedema appears to be through BK. Both histamine and BK are potent inflammatory mediators. However, the treatments for histamine-mediated angioedema are unsuitable for BK-mediated angioedema. We hypothesized that long-term consumption of tea would reduce bradykinin-dependent processes within the systemic and pulmonary vasculature, independent of the anti-inflammatory actions of polyphenols. A purified fraction of the black tea water extract inhibited both kallikrein and activated FXII. The black tea water extracts inhibited factor XII-induced cell migration and inhibited the production of kallikrein on the endothelial cell line. We compared the inhibitory effects of the black tea water extract and twenty-three well-known anti-inflammatory medicinal herbs, in inhibiting both kallikrein and FXII. Surprisingly, arjunglucoside II specifically inhibited the activated factor XII (FXIIa), but not the kallikrein and the activated factor XI. Taken together, the black tea water extract exerts its anti-inflammatory effects, in part, by inhibiting kallikrein and activated FXII, which are part of the plasma kallikrein-kinin system (KKS), and by decreasing BK production. The inhibition of kallikrein and activated FXII represents a unique polyphenol-independent anti-inflammatory mechanism of action for the black tea.


Asunto(s)
Bradiquinina/metabolismo , Camellia/química , Endotelio Vascular/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Sistema Calicreína-Quinina/efectos de los fármacos , Extractos Vegetales/farmacología , Arteria Pulmonar/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Arteria Pulmonar/metabolismo
2.
Biomaterials ; 272: 120778, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33812214

RESUMEN

Blood contact with high surface area medical devices, such as dialysis and extracorporeal life support (ECLS), induces rapid surface coagulation. Systemic anticoagulation, such as heparin, is thus necessary to slow clot formation, but some patients suffer from bleeding complications. Both problems might be reduced by 1) replacing heparin anticoagulation with artificial surface inhibition of the protein adsorption that initiates coagulation and 2) selective inhibition of the intrinsic branch of the coagulation cascade. This approach was evaluated by comparing clot formation and bleeding times during short-term ECLS using zwitterionic polycarboxybetaine (PCB) surface coatings combined with either a potent, selective, bicyclic peptide inhibitor of activated Factor XII (FXII900) or standard heparin anticoagulation. Rabbits underwent venovenous ECLS with small sham oxygenators for 60 min using three means of anticoagulation (n = 4 ea): (1) PCB coating + FXII900 infusion, (2) PCB coating + heparin infusion with an activated clotting time of 220-300s, and (3) heparin infusion alone. Sham oxygenator blood clot weights in the PCB + FXII900 and PCB + heparin groups were 4% and 25% of that in the heparin group (p < 10-6 and p < 10-5), respectively. At the same time, the bleeding time remained normal in the PCB + FXII900 group (2.4 ± 0.2 min) but increased to 4.8 ± 0.5 and 5.1 ± 0.7 min in the PCB + heparin and heparin alone groups (p < 10-4 and 0.01). Sham oxygenator blood flow resistance was significantly lower in the PCB + FXII900 and PCB + heparin groups than in the heparin only group (p < 10-6 and 10-5). These results were confirmed by gross and scanning electron microscopy (SEM) images and fibrinopeptide A (FPA) concentrations. Thus, the combined use of PCB coating and FXII900 markedly reduced sham oxygenator coagulation and tissue bleeding times versus the clinical standard of heparin anticoagulation and is a promising anticoagulation method for clinical ECLS.


Asunto(s)
Anticoagulantes/farmacología , Oxigenación por Membrana Extracorpórea , Factor XII/antagonistas & inhibidores , Animales , Betaína , Coagulación Sanguínea , Heparina/farmacología , Humanos , Ácidos Polimetacrílicos , Conejos , Diálisis Renal
3.
Clin Sci (Lond) ; 134(9): 1049-1061, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32309850

RESUMEN

Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.


Asunto(s)
Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Factor XII/antagonistas & inhibidores , Proteína ADAM17/metabolismo , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Modelos Animales de Enfermedad , Factor XII/metabolismo , Ratones
4.
Nat Commun ; 8: 16092, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28714475

RESUMEN

The rapid renal clearance of peptides in vivo limits this attractive platform for the treatment of a broad range of diseases that require prolonged drug half-lives. An intriguing approach for extending peptide circulation times works through a 'piggy-back' strategy in which peptides bind via a ligand to the long-lived serum protein albumin. In accordance with this strategy, we developed an easily synthesized albumin-binding ligand based on a peptide-fatty acid chimera that has a high affinity for human albumin (Kd=39 nM). This ligand prolongs the elimination half-life of cyclic peptides in rats 25-fold to over seven hours. Conjugation to a peptide factor XII inhibitor developed for anti-thrombotic therapy extends the half-life from 13 minutes to over five hours, inhibiting coagulation for eight hours in rabbits. This high-affinity albumin ligand could potentially extend the half-life of peptides in human to several days, substantially broadening the application range of peptides as therapeutics.


Asunto(s)
Portadores de Fármacos/química , Ácidos Grasos/química , Péptidos/administración & dosificación , Albúmina Sérica Humana/química , Acilación , Animales , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Factor XII/antagonistas & inhibidores , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacocinética , Semivida , Humanos , Ligandos , Tasa de Depuración Metabólica , Péptidos/metabolismo , Péptidos/farmacocinética , Péptidos Cíclicos/administración & dosificación , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacocinética , Calicreína Plasmática/antagonistas & inhibidores , Unión Proteica , Conejos , Ratas , Albúmina Sérica Humana/metabolismo , Albúmina Sérica Humana/farmacocinética , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
5.
Thromb Haemost ; 117(7): 1283-1288, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28594426

RESUMEN

Thrombosis is a leading cause of death and disability worldwide, and anticoagulants are the mainstay of its prevention and treatment. Starting with unfractionated heparin (UFH) and vitamin K antagonists (VKAs) such as warfarin, the choices of anticoagulants have exploded in the past 20 years. With over 90 % subcutaneous bioavailability, no need for coagulation monitoring and dose adjustment, and a lower risk of heparin-induced thrombocytopenia, low-molecular-weight heparin and fondaparinux have replaced UFH for prevention and initial treatment of venous thromboembolism and for secondary prevention in cancer patients. In patients undergoing percutaneous interventions, bivalirudin is often used instead of UFH. Oral anticoagulation therapy has advanced with the introduction of the non-vitamin K antagonist oral anticoagulants (NOACs), which include dabigatran, rivaroxaban, apixaban and edoxaban. With efficacy at least equal to that of VKAs but with greater safety and convenience, the NOACs are now replacing VKAs for many indications. This paper a) highlights these advances, b) outlines how specific reversal agents for the NOACs will enhance their safety, c) reviews some of the ongoing trials with the NOACs, and d) describes the inhibitors of factor XII and XI that are under investigation as anticoagulants.


Asunto(s)
Anticoagulantes/uso terapéutico , Anticoagulantes/administración & dosificación , Anticoagulantes/efectos adversos , Antídotos/uso terapéutico , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Descubrimiento de Drogas , Factor XI/antagonistas & inhibidores , Factor XII/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Heparina/efectos adversos , Humanos , Enfermedad Arterial Periférica/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico , Trombosis/sangre , Trombosis/tratamiento farmacológico , Trombosis/prevención & control , Tromboembolia Venosa/tratamiento farmacológico , Vitamina K/antagonistas & inhibidores , Warfarina/efectos adversos
6.
J Med Chem ; 60(3): 1151-1158, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28045547

RESUMEN

Factor XII (FXII) is a plasma protease that has emerged in recent years as a potential target to treat or prevent pathological thrombosis, to inhibit contact activation in extracorporeal circulation, and to treat the swelling disorder hereditary angioedema. While several protein based inhibitors with high affinity for activated FXII (FXIIa) were developed, the generation of small molecule inhibitors has been challenging. In this work, we have generated a potent and selective FXIIa inhibitor by optimizing a peptide macrocycle that was recently evolved by phage display (Ki = 0.84 ± 0.03 nM). A fluorine atom introduced in the para-position of phenylalanine enhanced the binding affinity as much as 10-fold. Furthermore, we improved the proteolytic stability by substituting the N-terminal arginine by norarginine. The resulting inhibitor combines high inhibitory affinity and selectivity with a good stability in plasma (Ki = 1.63 ± 0.18 nM, >27 000-fold selectivity, t1/2(plasma) =16 ± 4 h). The inhibitor efficiently blocked activation of the intrinsic coagulation pathway in human blood ex vivo.


Asunto(s)
Factor XII/antagonistas & inhibidores , Compuestos Macrocíclicos/farmacología , Péptidos/química , Humanos , Compuestos Macrocíclicos/química , Proteolisis
7.
Ann Biomed Eng ; 45(5): 1328-1340, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27933406

RESUMEN

In the contact activation pathway of the coagulation, zymogen factor XII (FXII) is converted to FXIIa, which triggers activation of FXI leading to the activation of FIX and subsequent thrombin generation and fibrin formation. Feedback activation of FXI by thrombin has been shown to promote thrombin generation in a FXII-independent manner and FXIIa can bypass FXI to directly activate FX and prothrombin in the presence of highly negatively charged molecules, such as long-chain polyphosphates (LC polyP). We sought to determine whether activation of FXII or FXI differentially regulate the physical biology of fibrin formation. Fibrin formation was initiated with tissue factor, ellagic acid (EA), or LC polyP in the presence of inhibitors of FXI and FXII. Our data demonstrated that inhibition of FXI decreased the rate of fibrin formation and fiber network density, and increased the fibrin network strength and rate of fibrinolysis when gelation was initiated via the contact activation pathway with EA. FXII inhibition decreased the fibrin formation and fibrin density, and increased the fibrinolysis rate only when fibrin formation was initiated via the contact activation pathway with LC polyP. Overall, we demonstrate that inhibition of FXI and FXII distinctly alter the biophysical properties of fibrin.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/química , Factor XII , Factor XI , Fibrina/química , Fibrinólisis , Factor XI/antagonistas & inhibidores , Factor XI/química , Factor XII/antagonistas & inhibidores , Factor XII/química , Humanos , Polifosfatos/química
8.
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
9.
Blood ; 129(2): 147-154, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-27780803

RESUMEN

Despite the introduction of direct oral anticoagulants (DOACs), the search for more effective and safer antithrombotic strategies continues. Better understanding of the pathogenesis of thrombosis has fostered 2 new approaches to achieving this goal. First, evidence that thrombin may be as important as platelets to thrombosis at sites of arterial injury and that platelets contribute to venous thrombosis has prompted trials comparing anticoagulants with aspirin for secondary prevention in arterial thrombosis and aspirin with anticoagulants for primary and secondary prevention of venous thrombosis. These studies will help identify novel treatment strategies. Second, emerging data that naturally occurring polyphosphates activate the contact system and that this system is critical for thrombus stabilization and growth have identified factor XII (FXII) and FXI as targets for new anticoagulants that may be even safer than the DOACs. Studies are needed to determine whether FXI or FXII is the better target and to compare the efficacy and safety of these new strategies with current standards of care for the prevention or treatment of thrombosis. Focusing on these advances, this article outlines how treatment strategies for thrombosis are evolving and describes the rationale and approaches to targeting FXII and FXI. These emerging anticoagulant strategies should address unmet needs and reduce the systemic underuse of anticoagulation because of the fear of bleeding.


Asunto(s)
Anticoagulantes/farmacología , Coagulación Sanguínea/fisiología , Trombosis/metabolismo , Animales , Coagulación Sanguínea/efectos de los fármacos , Factor XI/antagonistas & inhibidores , Factor XII/antagonistas & inhibidores , Humanos
10.
Thromb Res ; 141 Suppl 2: S4-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27207422

RESUMEN

Cancer is an established risk factor for venous thromboembolism (VTE) and VTE is the second leading cause of death in patients with cancer. The incidence of cancer-related thrombosis is rising and is associated with worse outcomes. Despite our growing understanding on tumor-driven procoagulant mechanisms including cancer-released procoagulant proteases, expression of tissue factor on cancer cells and derived microvesicles, as well as alterations in the extracellular matrix of the cancer cell milieu, anticoagulation therapy in cancer patients has remained challenging. This review comments on a newly discovered cancer-associated procoagulant pathway. Experimental VTE models in mice and studies on patient cancer material revealed that prostate cancer cells and associated exosomes display the inorganic polymer polyphosphate on their plasma membrane. Polyphosphate activates blood coagulation factor XII and initiates thrombus formation via the intrinsic pathway of coagulation. Pharmacologic inhibition of factor XII activity protects mice from VTE and reduces thrombin coagulant activity in plasma of prostate cancer patients. Factor XII inhibitors provide thrombo-protection without impairing hemostatic mechanisms and thus, unlike currently used anticoagulants, do not increase bleeding risk. Interference with the polyphosphate/factor XII pathway may provide the novel opportunity for safe anticoagulation therapy in patients with malignancies.


Asunto(s)
Anticoagulantes/uso terapéutico , Factor XII/metabolismo , Neoplasias/complicaciones , Polifosfatos/metabolismo , Trombosis/complicaciones , Trombosis/tratamiento farmacológico , Animales , Anticoagulantes/efectos adversos , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Humanos , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Embolia Pulmonar/complicaciones , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/metabolismo , Trombosis/sangre , Trombosis/metabolismo , Tromboembolia Venosa/complicaciones , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/metabolismo
11.
Thromb Res ; 141 Suppl 2: S40-5, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27207423

RESUMEN

Although the non-vitamin antagonist oral anticoagulants produce less intracranial bleeding than warfarin, serious bleeding still occurs. Therefore, the search for safer anticoagulants continues. Factor XII and factor XI have emerged as promising targets whose inhibition has the potential to prevent thrombosis with little or no disruption of hemostasis. Thus, thrombosis is attenuated in mice deficient in factor XII or factor XI and patients with congenital factor XII deficiency do not bleed and those with factor XI deficiency rarely have spontaneous bleeding. Strategies targeting factor XII and XI include antisense oligonucleotides to decrease their synthesis, inhibitory antibodies or aptamers, and small molecule inhibitors. These strategies attenuate thrombosis in various animal models and factor XI knockdown with an antisense oligonucleotide in patients undergoing knee replacement surgery reduced postoperative venous thromboembolism to a greater extent than enoxaparin without increasing bleeding. Therefore, current efforts are focused on evaluating the efficacy and safety of factor XII and factor XI directed anticoagulant strategies.


Asunto(s)
Anticoagulantes/uso terapéutico , Descubrimiento de Drogas , Factor XII/antagonistas & inhibidores , Factor XI/antagonistas & inhibidores , Terapia Molecular Dirigida , Trombosis/tratamiento farmacológico , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Anticoagulantes/farmacología , Aptámeros de Nucleótidos/farmacología , Aptámeros de Nucleótidos/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Descubrimiento de Drogas/métodos , Factor XI/metabolismo , Factor XII/metabolismo , Humanos , Terapia Molecular Dirigida/métodos , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Trombosis/sangre , Trombosis/metabolismo
12.
J Thromb Haemost ; 13(8): 1383-95, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25976012

RESUMEN

The most commonly used anticoagulants produce therapeutic antithrombotic effects either by inhibiting thrombin or factor Xa (FXa) or by lowering the plasma levels of the precursors of these key enzymes, prothrombin and FX. These drugs do not distinguish between thrombin generation contributing to thrombosis from thrombin generation required for hemostasis. Thus, anticoagulants increase bleeding risk, and many patients who would benefit from therapy go untreated because of comorbidities that place them at unacceptable risk for hemorrhage. Studies in animals demonstrate that components of the plasma contact activation system contribute to experimentally induced thrombosis, despite playing little or no role in hemostasis. Attention has focused on FXII, the zymogen of a protease (FXIIa) that initiates contact activation when blood is exposed to foreign surfaces, and FXI, the zymogen of the protease FXIa, which links contact activation to the thrombin generation mechanism. In the case of FXI, epidemiologic data indicate this protein contributes to stroke and venous thromboembolism, and perhaps myocardial infarction, in humans. A phase 2 trial showing that reduction of FXI may be more effective than low molecular weight heparin at preventing venous thrombosis during knee replacement surgery provides proof of concept for the premise that an antithrombotic effect can be uncoupled from an anticoagulant effect in humans by targeting components of contact activation. Here, we review data on the role of FXI and FXII in thrombosis and results of preclinical and human trials for therapies targeting these proteins.


Asunto(s)
Anticoagulantes/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Factor XI/antagonistas & inhibidores , Fibrinolíticos/uso terapéutico , Trombosis de la Vena/tratamiento farmacológico , Animales , Anticoagulantes/efectos adversos , Modelos Animales de Enfermedad , Diseño de Fármacos , Activación Enzimática , Factor XI/metabolismo , Factor XII/metabolismo , Fibrinolíticos/efectos adversos , Hemorragia/inducido químicamente , Humanos , Terapia Molecular Dirigida , Factores de Riesgo , Resultado del Tratamiento , Trombosis de la Vena/sangre , Trombosis de la Vena/diagnóstico
13.
Eur J Med Chem ; 92: 257-69, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25559206

RESUMEN

Fucosylated chondroitin sulfate (FCS), a structurally unusual glycosaminoglycan, has distinct anticoagulant properties, and is an especially strong inhibitor of the intrinsic factor Xase (anti-Xase). To obtain a highly selective inhibitor of human Xase, we purified six native FCSs with various sulfation patterns, prepared a series of FCS derivatives, and then elucidated the relationship between the structures and the anticoagulant activities of FCSs. FCSs 1-3 containing higher Fuc2S4S exhibit stronger AT-dependent anti-IIa activities, whereas 4-6 containing more Fuc3S4S produce potent HCII-dependent anti-IIa activities. Saccharides containing a minimum of 6-8 trisaccharide units, free carboxyl groups, and full fucosylation of GlcA may be required for potent anti-Xase activity, and approximately six trisaccharide units and partial fucosylation of GlcA may contribute to potent HCII-dependent activity. Decreasing of the molecular weights markedly reduces their AT-dependent anti-IIa activities, and even eliminates human platelet and factor XII activation. Furthermore, in vitro and in vivo studies suggested that fractions of 6-12 kDa may be very promising compounds as putative selective intrinsic Xase inhibitors with antithrombotic action, but without the consequences of major bleeding and factor XII activation.


Asunto(s)
Anticoagulantes/farmacología , Sulfatos de Condroitina/farmacología , Cisteína Endopeptidasas/metabolismo , Inhibidores Enzimáticos/farmacología , Factor XII/antagonistas & inhibidores , Fibrinolíticos/farmacología , Animales , Anticoagulantes/síntesis química , Anticoagulantes/química , Coagulación Sanguínea/efectos de los fármacos , Sulfatos de Condroitina/síntesis química , Sulfatos de Condroitina/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Factor XII/metabolismo , Fibrinolíticos/síntesis química , Fibrinolíticos/química , Humanos , Ratones , Ratones Endogámicos , Conformación Molecular , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Trombosis/tratamiento farmacológico
14.
J Allergy Clin Immunol ; 135(4): 1031-1043.e6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25240785

RESUMEN

BACKGROUND: Anaphylaxis is an acute, potentially lethal, multisystem syndrome resulting from the sudden release of mast cell-derived mediators into the circulation. OBJECTIVES AND METHODS: We report here that a plasma protease cascade, the factor XII-driven contact system, critically contributes to the pathogenesis of anaphylaxis in both murine models and human subjects. RESULTS: Deficiency in or pharmacologic inhibition of factor XII, plasma kallikrein, high-molecular-weight kininogen, or the bradykinin B2 receptor, but not the B1 receptor, largely attenuated allergen/IgE-mediated mast cell hyperresponsiveness in mice. Reconstitutions of factor XII null mice with human factor XII restored susceptibility for allergen/IgE-mediated hypotension. Activated mast cells systemically released heparin, which provided a negatively charged surface for factor XII autoactivation. Activated factor XII generates plasma kallikrein, which proteolyzes kininogen, leading to the liberation of bradykinin. We evaluated the contact system in patients with anaphylaxis. In all 10 plasma samples immunoblotting revealed activation of factor XII, plasma kallikrein, and kininogen during the acute phase of anaphylaxis but not at basal conditions or in healthy control subjects. The severity of anaphylaxis was associated with mast cell degranulation, increased plasma heparin levels, the intensity of contact system activation, and bradykinin formation. CONCLUSIONS: In summary, the data collectively show a role of the contact system in patients with anaphylaxis and support the hypothesis that targeting bradykinin generation and signaling provides a novel and alternative treatment strategy for anaphylactic attacks.


Asunto(s)
Anafilaxia/inmunología , Anafilaxia/metabolismo , Factor XII/metabolismo , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Mastocitos/inmunología , Adulto , Anciano , Anafilaxia/complicaciones , Anafilaxia/genética , Animales , Biomarcadores , Bradiquinina/metabolismo , Modelos Animales de Enfermedad , Factor XII/antagonistas & inhibidores , Factor XII/genética , Femenino , Humanos , Hipersensibilidad/complicaciones , Hipersensibilidad/genética , Hipotensión/etiología , Quininógenos/metabolismo , Masculino , Ratones Noqueados , Persona de Mediana Edad , Receptor de Bradiquinina B2/genética , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal , Factores de Tiempo , Adulto Joven
15.
PLoS Pathog ; 9(7): e1003470, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874198

RESUMEN

Hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS) are diseases caused by hantavirus infections and are characterized by vascular leakage due to alterations of the endothelial barrier. Hantavirus-infected endothelial cells (EC) display no overt cytopathology; consequently, pathogenesis models have focused either on the influx of immune cells and release of cytokines or on increased degradation of the adherens junction protein, vascular endothelial (VE)-cadherin, due to hantavirus-mediated hypersensitization of EC to vascular endothelial growth factor (VEGF). To examine endothelial leakage in a relevant in vitro system, we co-cultured endothelial and vascular smooth muscle cells (vSMC) to generate capillary blood vessel-like structures. In contrast to results obtained in monolayers of cultured EC, we found that despite viral replication in both cell types as well as the presence of VEGF, infected in vitro vessels neither lost integrity nor displayed evidence of VE-cadherin degradation. Here, we present evidence for a novel mechanism of hantavirus-induced vascular leakage involving activation of the plasma kallikrein-kinin system (KKS). We show that incubation of factor XII (FXII), prekallikrein (PK), and high molecular weight kininogen (HK) plasma proteins with hantavirus-infected EC results in increased cleavage of HK, higher enzymatic activities of FXIIa/kallikrein (KAL) and increased liberation of bradykinin (BK). Measuring cell permeability in real-time using electric cell-substrate impedance sensing (ECIS), we identified dramatic increases in endothelial cell permeability after KKS activation and liberation of BK. Furthermore, the alterations in permeability could be prevented using inhibitors that directly block BK binding, the activity of FXIIa, or the activity of KAL. Lastly, FXII binding and autoactivation is increased on the surface of hantavirus-infected EC. These data are the first to demonstrate KKS activation during hantavirus infection and could have profound implications for treatment of hantavirus infections.


Asunto(s)
Capilares/virología , Permeabilidad Capilar , Endotelio Vascular/virología , Activación Enzimática , Factor XII/metabolismo , Infecciones por Hantavirus/virología , Sistema Calicreína-Quinina , Bradiquinina/antagonistas & inhibidores , Bradiquinina/metabolismo , Capilares/efectos de los fármacos , Capilares/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Orthohantavirus/fisiología , Infecciones por Hantavirus/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Sistema Calicreína-Quinina/efectos de los fármacos , Quininógeno de Alto Peso Molecular/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/virología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/virología , Precalicreína/antagonistas & inhibidores , Precalicreína/metabolismo , Inhibidores de Proteasas/farmacología , Proteolisis/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/virología , Propiedades de Superficie , Replicación Viral
16.
J Obstet Gynaecol Res ; 39(7): 1223-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23803004

RESUMEN

Factor XII, plasma prekallikrein and high-molecular-weight kininogen were first identified as coagulation proteins in the intrinsic pathway because patients deficient in these proteins had marked prolongation of in vitro surface-activated coagulation time. However, deficiencies of these proteins are not associated with clinical bleeding. Paradoxically, studies suggest that these proteins have anticoagulant and profibrinolytic activities. In fact, association between deficiencies of these proteins and thrombosis has been reported. Recently, autoantibodies to these proteins and antiphospholipid antibodies are frequent coagulation-related abnormalities found in unexplained recurrent aborters. Evidence has accumulated for the presence of the kallikrein-kininogen-kinin system in the fetoplacental unit. The contact system, or kallikrein-kininogen-kinin system, in the reproductive tract plays an essential roll in the regulation of thrombosis, hemostasis, angiogenesis and in the defense against invasive bacterial infection. Autoantibodies to these proteins may be associated with pregnancy losses due to disruption of this system. These possibilities will be reviewed, the functions of the individual components will be summarized, and their role in blood coagulation and pregnancy discussed.


Asunto(s)
Aborto Habitual/etiología , Anticuerpos Antifosfolípidos/análisis , Autoanticuerpos/análisis , Enfermedades Autoinmunes/fisiopatología , Factor XII/antagonistas & inhibidores , Quininógenos/metabolismo , Fosfatidiletanolaminas/antagonistas & inhibidores , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Factor XII/metabolismo , Femenino , Humanos , Fosfatidiletanolaminas/metabolismo , Embarazo
17.
Thromb Haemost ; 110(2): 223-32, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23739841

RESUMEN

Antithrombotic drugs like vitamin K antagonists and heparin have been the gold standard for the treatment and prevention of thromboembolic disease for many years. Unfortunately, there are several disadvantages of these antithrombotic drugs: they are accompanied by serious bleeding problems, it is necessary to monitor the therapeutic window, and there are various interactions with food and other drugs. This has led to the development of new oral anticoagulants, specifically inhibiting either thrombin or factor Xa. In terms of effectiveness, these drugs are comparable to the currently available anticoagulants; however, they are still associated with issues such as bleeding, reversal of the drug and complicated laboratory monitoring. Vitamin K antagonists, heparin, direct thrombin and factor Xa inhibitors have in common that they target key proteins of the haemostatic system. In an attempt to overcome these difficulties we investigated whether the intrinsic coagulation factors (VIII, IX, XI, XII, prekallikrein and high-molecular-weight kininogen) are superior targets for anticoagulation. We analysed epidemiological data concerning thrombosis and bleeding in patients deficient in one of the intrinsic pathway proteins. Furthermore, we discuss several thrombotic models in intrinsic coagulation factor-deficient animals. The combined results suggest that intrinsic coagulation factors could be suitable targets for anticoagulant drugs.


Asunto(s)
Anticoagulantes/uso terapéutico , Inhibidores del Factor Xa , Trombina/antagonistas & inhibidores , Administración Oral , Animales , Anticoagulantes/administración & dosificación , Anticoagulantes/efectos adversos , Factores de Coagulación Sanguínea/antagonistas & inhibidores , Diseño de Fármacos , Factor IX/antagonistas & inhibidores , Factor VIII/antagonistas & inhibidores , Factor XI/antagonistas & inhibidores , Deficiencia del Factor XI/complicaciones , Factor XII/antagonistas & inhibidores , Hemorragia/inducido químicamente , Hemostasis/efectos de los fármacos , Humanos , Inflamación/sangre , Quininógenos/antagonistas & inhibidores , Precalicreína/antagonistas & inhibidores , Accidente Cerebrovascular/prevención & control , Trombosis/sangre , Trombosis/tratamiento farmacológico , Trombosis/prevención & control
18.
Adv Exp Med Biol ; 491: 343-50, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-14533807

RESUMEN

It has long been controversial whether sulfatide stimulates blood coagulation or inhibits blood coagulation. In this paper, I demonstrated not only anticoagulant activity but also coagulant activity of sulfatide in vivo by using experimental animal models and presented the possibility that sulfatide may function contradictorily under physiological and/or pathological conditions. For instance, I presented possible involvement of sulfatide in thrombotic diseases associated with cancers.


Asunto(s)
Coagulación Sanguínea/fisiología , Sulfoglicoesfingolípidos/sangre , Animales , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Factor XII/antagonistas & inhibidores , Humanos , Técnicas In Vitro , Ratones , Neoplasias/sangre , Neoplasias/complicaciones , Ratas , Sulfoglicoesfingolípidos/farmacología , Trombosis/sangre , Trombosis/etiología
19.
Blood ; 87(6): 2337-44, 1996 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-8630396

RESUMEN

In previous studies, we have shown that administration of monoclonal antibody (MoAb) C6B7 against human factor XII to baboons challenged with a lethal dose of Escherichia coli abrogates activation of the contact system and modulates secondary hypotension. To evaluate the contribution of activated contact proteases to the appearance of other inflammatory mediators in this experimental model of sepsis, we studied the effect of administration of MoAb C6B7 on activation of complement and fibrinolytic cascades, stimulation of neutrophil degranulation, and release of the proinflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6). Activation of the complement system, as reflected by circulating C3b/c and C4b/c levels, was significantly reduced in five animals that had received MoAb C6B7 before a lethal dose of E coli as compared with five control animals that had been given a lethal challenge only. Inhibition of contact activation also modulated the fibrinolytic response, since the release of tissue-type plasminogen activator (t-PA) and the appearance of plasmin-alpha2-antiplasmin (PAP) complexes into the circulation was significantly attenuated upon pretreatment with anti-factor XII MoAb. In contrast, plasma levels of plasminogen activator inhibitor (PAI) were modestly enhanced in the treatment group. Degranulation of neutrophils, as assessed by circulating elastase-alpha1-protease inhibitor complexes, and release of IL-6 but not of TNF-alpha was decreased in anti-factor XII-treated animals. Observed differences in the inflammatory response between treatment and control groups were not likely due to different challenges, since the number of E coli that had been infused, as well as circulating levels of endotoxin after the challenge, were similar for both groups. These data suggest that activation of the contact system modulates directly or indirectly various mediator systems involved in the inflammatory response during severe sepsis in nonhuman primates.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Bacteriemia/sangre , Activación de Complemento/efectos de los fármacos , Infecciones por Escherichia coli/sangre , Factor XII/antagonistas & inhibidores , Fibrinólisis/efectos de los fármacos , Interleucina-6/metabolismo , Elastasa Pancreática/metabolismo , Choque Séptico/sangre , Animales , Anticuerpos Monoclonales/inmunología , Bacteriemia/terapia , Citocinas/metabolismo , Infecciones por Escherichia coli/terapia , Factor XII/inmunología , Factor XII/fisiología , Fibrinolisina/análisis , Inmunización Pasiva , Elastasa de Leucocito , Ratones , Neutrófilos/enzimología , Neutrófilos/metabolismo , Papio , Inhibidor 1 de Activador Plasminogénico/metabolismo , Choque Séptico/terapia , Activador de Tejido Plasminógeno/metabolismo , alfa 2-Antiplasmina/análisis
20.
Thromb Haemost ; 73(5): 798-804, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7482406

RESUMEN

Lupus anticoagulants are a group of antibodies commonly found in patients with autoimmune diseases such as systemic lupus erythematosus. Lupus anticoagulants inhibit phospholipid dependent coagulation and may bind to negatively charged phospholipids. Recent studies have suggested an association between anti-beta 2-glycoprotein I and a lupus anticoagulant, whose activity is frequently dependent on the presence of beta 2-glycoprotein I. Based on these observations, the effect of anti-beta 2-glycoprotein I on the autoactivation of factor XII in plasma was investigated. Autoactivation initiated by the presence of negatively charged phospholipids, but not by sulfatide, was strongly inhibited by immunoaffinity purified anti-beta 2-glycoprotein I. The dose-response curve of anti-beta 2-glycoprotein I was identical with that of a precipitating antibody, showing no inhibition at low and high antibody dilutions and maximal inhibition at an intermediate dilution. At high antibody concentrations, an increased rate of factor XIIa activation was observed. This increase was of the same magnitude as the decreased rate observed in plasma supplemented with the same amount of beta 2-glycoprotein I as in the plasma itself. This confirms the inhibitory effect of beta 2-GP-I on the contact activation and shows that inhibition is effective on the autoactivation of factor XII in plasma. The inhibitory action of beta 2-glycoprotein I was independent of the inhibition caused by the anti-beta 2-glycoprotein I/beta 2 glycoprotein I complex suggesting a synchronized inhibition of factor XII autoactivation by beta 2-glycoprotein I and anti-beta 2-glycoprotein I. The inhibition caused by the antibody is suggested to be caused by a reduced availability of negatively charged phospholipids due to the binding of the anti-beta 2-GP-I/beta 2-GP-I complex. This complex may be a lupus anticoagulant.


Asunto(s)
Autoanticuerpos/farmacología , Factor XII/metabolismo , Factor XIIa/análisis , Glicoproteínas/farmacología , Fosfolípidos/antagonistas & inhibidores , Animales , Anticuerpos Antiidiotipos/inmunología , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/inmunología , Compuestos Cromogénicos , Relación Dosis-Respuesta Inmunológica , Activación Enzimática/efectos de los fármacos , Epítopos/inmunología , Factor XII/antagonistas & inhibidores , Factor XIIa/biosíntesis , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/inmunología , Humanos , Inhibidor de Coagulación del Lupus/inmunología , Inhibidor de Coagulación del Lupus/farmacología , Fosfatos de Fosfatidilinositol/farmacología , Fosfolípidos/farmacología , Unión Proteica , Conejos , Sulfoglicoesfingolípidos/farmacología , beta 2 Glicoproteína I
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA