Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Thromb Haemost ; 120(6): 883-993, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32375196

RESUMEN

Cardiovascular disease, including stroke, myocardial infarction, and venous thromboembolism, is one of the leading causes of morbidity and mortality worldwide. Excessive coagulation may cause vascular occlusion in arteries and veins eventually leading to thrombotic diseases. Studies in recent years suggest that coagulation factors are involved in these pathological mechanisms. Factors XIa (FXIa), XIIa (FXIIa), and plasma kallikrein (PKa) of the contact system of coagulation appear to contribute to thrombosis while playing a limited role in hemostasis. Contact activation is initiated upon autoactivation of FXII on negatively charged surfaces. FXIIa activates plasma prekallikrein (PK) to PKa, which in turn activates FXII and initiates the kallikrein-kinin pathway. FXI is also activated by FXIIa, leading to activation of FIX and finally to thrombin formation, which in turn activates FXI in an amplification loop. Animal studies have shown that arterial and venous thrombosis can be reduced by the inhibition of FXI(a) or PKa. Furthermore, data from human studies suggest that these enzymes may be valuable targets to reduce thrombosis risk. In this review, we discuss the structure and function of FXI(a) and PK(a), their involvement in the development of venous and arterial thrombosis in animal models and human studies, and current therapeutic strategies.


Asunto(s)
Arteriopatías Oclusivas/sangre , Factor XIa/fisiología , Calicreína Plasmática/fisiología , Trombosis/sangre , Animales , Coagulación Sanguínea/fisiología , Trastornos de la Coagulación Sanguínea/sangre , Factores de Coagulación Sanguínea/fisiología , Modelos Animales de Enfermedad , Activación Enzimática , Deficiencia del Factor XI/sangre , Factor XIa/química , Factor XIa/inmunología , Inhibidores del Factor Xa/uso terapéutico , Humanos , Ratones , Ratones Noqueados , Precalicreína/deficiencia , Precalicreína/metabolismo , Procesamiento Proteico-Postraduccional , Especificidad de la Especie , Trombofilia/tratamiento farmacológico , Trombosis de la Vena/sangre
2.
Semin Ophthalmol ; 31(1-2): 19-24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26959125

RESUMEN

Diabetic retinal disease is characterized by a series of retinal microvascular changes and increases in retinal vascular permeability that lead to development of diabetic retinopathy (DR) and diabetic macular edema (DME), respectively. Current treatment strategies for DR and DME are mostly limited to vascular endothelial growth factor (VEGF) inhibitors and laser photocoagulation. These treatment modalities are not universally effective in all patients, and potential side effects persist in a significant portion of patients. The plasma kallikrein-kinin system (KKS) is one of the pathways that has been identified in the vitreous in proliferative DR and DME. Preclinical studies have shown that the activation of intraocular KKS induces retinal vascular permeability, vasodilation, and retinal thickening. Proteomic analysis from vitreous of eyes with DME has shown that KKS and VEGF pathways are potentially independent biologic pathways. Furthermore, proteins associated with DME in the vitreous were significantly more correlated with the KKS pathway compared to VEGF pathway. Preclinical experiments on diabetic animals showed that inhibition of KKS components was found to be an effective approach to decrease retinal vascular permeability. An initial phase I human trial of a novel plasma kallikrein inhibitor for the treatment of DME is currently ongoing to test the safety of this approach and serves as an initial step in the translation of basic science discovery into an innovative clinical intervention.


Asunto(s)
Retinopatía Diabética/fisiopatología , Sistema Calicreína-Quinina/fisiología , Edema Macular/fisiopatología , Animales , Permeabilidad Capilar , Retinopatía Diabética/terapia , Humanos , Edema Macular/terapia , Calicreína Plasmática/fisiología , Vasos Retinianos/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
4.
Thromb Haemost ; 110(3): 434-41, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23676986

RESUMEN

Plasma kallikrein (PK) is a serine protease generated from plasma prekallikrein, an abundant circulating zymogen expressed by the Klkb1 gene. The physiological actions of PK have been primarily attributed to its production of bradykinin and activation of coagulation factor XII, which promotes inflammation and the intrinsic coagulation pathway. Recent genetic, molecular, and pharmacological studies of PK have provided further insight into its role in physiology and disease. Genetic analyses have revealed common Klkb1 variants that are association with blood metabolite levels, hypertension, and coagulation. Characterisation of animal models with Klkb1 deficiency and PK inhibition have demonstrated effects on inflammation, vascular function, blood pressure regulation, thrombosis, haemostasis, and metabolism. These reports have also identified a host of PK substrates and interactions, which suggest an expanded physiological role for this protease beyond the bradykinin system and coagulation. The review summarises the mechanisms that contribute to PK activation and its emerging role in diabetes and metabolism.


Asunto(s)
Diabetes Mellitus/sangre , Regulación de la Expresión Génica , Calicreína Plasmática/fisiología , Adipogénesis , Animales , Coagulación Sanguínea , Presión Sanguínea , Bradiquinina/metabolismo , Factor XII/metabolismo , Variación Genética , Glucosa/metabolismo , Hemostasis , Humanos , Hipertensión/metabolismo , Inflamación , Ratones , Precalicreína/metabolismo , Trombosis/metabolismo
5.
Nat Med ; 17(2): 206-10, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21258336

RESUMEN

Hyperglycemia is associated with greater hematoma expansion and poor clinical outcomes after intracerebral hemorrhage. We show that cerebral hematoma expansion triggered by intracerebral infusion of autologous blood is greater in diabetic rats and mice compared to nondiabetic controls and that this augmented expansion is ameliorated by plasma kallikrein (PK) inhibition or deficiency. Intracerebral injection of purified PK augmented hematoma expansion in both diabetic and acutely hyperglycemic rats, whereas injection of bradykinin, plasmin or tissue plasminogen activator did not elicit such a response. This response, which occurs rapidly, was prevented by co-injection of the glycoprotein VI agonist convulxin and was mimicked by glycoprotein VI inhibition or deficiency, implicating an effect of PK on inhibiting platelet aggregation. We show that PK inhibits collagen-induced platelet aggregation by binding collagen, a response enhanced by elevated glucose concentrations. The effect of hyperglycemia on hematoma expansion and PK-mediated inhibition of platelet aggregation could be mimicked by infusing mannitol. These findings suggest that hyperglycemia augments cerebral hematoma expansion by PK-mediated osmotic-sensitive inhibition of hemostasis.


Asunto(s)
Hemorragia Cerebral/fisiopatología , Hematoma/fisiopatología , Hiperglucemia/fisiopatología , Calicreína Plasmática/fisiología , Animales , Barrera Hematoencefálica/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Diabetes Mellitus Experimental/fisiopatología , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Calicreína Plasmática/farmacología , Plasminógeno/fisiología , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Ratas , Ratas Sprague-Dawley
6.
Curr Drug Targets ; 6(5): 551-7, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16026275

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. Also, deficiencies of these proteins, auto-antibodies to these proteins and anti-phospholipid antibodies are frequent hemostatis-related abnormalities found in unexplained recurrent aborters. Recently, evidence has accumulated for the presence of the kallikrein-kininogen-kinin system in the fetoplacental unit. Since contact proteins or kallikrein-kininogen-kinin system may play an important role in pregnancy especially in the fetoplacental unit, deficiencies of these proteins and/or auto-antibodies to these proteins may be associated with pregnancy losses. 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)
Factor XII/fisiología , Quininógenos/fisiología , Complicaciones del Embarazo/fisiopatología , Precalicreína/fisiología , Animales , Femenino , Humanos , Quininógeno de Alto Peso Molecular/fisiología , Calicreína Plasmática/fisiología , Embarazo , Complicaciones Hematológicas del Embarazo/terapia
7.
Biochem J ; 381(Pt 3): 841-6, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15117283

RESUMEN

Human selenoprotein P (SeP), a selenium-rich plasma glycoprotein, is presumed to contain ten selenocysteine residues; one of which is located at the 40th residue in the N-terminal region and the remaining nine localized in the C-terminal third part. We have shown that SeP not only catalyses the reduction of phosphatidylcholine hydroperoxide by glutathione [Saito, Hayashi, Tanaka, Watanabe, Suzuki, Saito and Takahashi (1999) J. Biol. Chem. 274, 2866-2871], but also supplies its selenium to proliferating cells [Saito and Takahashi (2002) Eur. J. Biochem. 269, 5746-5751]. Treatment of SeP with plasma kallikrein resulted in a sequential limited proteolysis (Arg-235-Gln-236 and Arg-242-Asp-243). The N-terminal (residues 1-235) and C-terminal (residues 243-361) fragments exhibited enzyme activity and selenium-supply activity respectively. These results confirm that SeP is a bi-functional protein and suggest that the first selenocysteine residue is the active site of the enzyme and the remaining nine residues function as a selenium supplier.


Asunto(s)
Péptidos/química , Proteínas/química , Sitios de Unión , Coagulación Sanguínea/fisiología , Línea Celular Tumoral , Glutatión Peroxidasa/metabolismo , Humanos , Hidrólisis , Células Jurkat/enzimología , Células Jurkat/metabolismo , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Calicreína Plasmática/metabolismo , Calicreína Plasmática/fisiología , Estructura Terciaria de Proteína , Proteínas/metabolismo , Selenio/metabolismo , Selenoproteína P , Selenoproteínas , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
8.
Braz J Med Biol Res ; 36(8): 1055-9, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12886459

RESUMEN

Human plasma kallikrein, a serine proteinase, plays a key role in intrinsic blood clotting, in the kallikrein-kinin system, and in fibrinolysis. The proteolytic enzymes involved in these processes are usually controlled by specific inhibitors and may be influenced by several factors including glycosaminoglycans, as recently demonstrated by our group. The aim of the present study was to investigate the effect of glycosaminoglycans (30 to 250 micro/ml) on kallikrein activity on plasminogen and factor XII and on the inhibition of kallikrein by the plasma proteins C1-inhibitor and antithrombin. Almost all available glycosaminoglycans (heparin, heparan sulfate, bovine and tuna dermatan sulfate, chondroitin 4- and 6-sulfates) reduced (1.2 to 3.0 times) the catalytic efficiency of kallikrein (in a nanomolar range) on the hydrolysis of plasminogen (0.3 to 1.8 microM) and increased (1.9 to 7.7 times) the enzyme efficiency in factor XII (0.1 to 10 microM) activation. On the other hand, heparin, heparan sulfate, and bovine and tuna dermatan sulfate improved (1.2 to 3.4 times) kallikrein inhibition by antithrombin (1.4 microM), while chondroitin 4- and 6-sulfates reduced it (1.3 times). Heparin and heparan sulfate increased (1.4 times) the enzyme inhibition by the C1-inhibitor (150 nM).


Asunto(s)
Factor XII/efectos de los fármacos , Fibrinolíticos/farmacología , Glicosaminoglicanos/farmacología , Calicreína Plasmática/efectos de los fármacos , Plasminógeno/efectos de los fármacos , Animales , Bovinos , Proteínas Inactivadoras del Complemento 1/efectos de los fármacos , Proteína Inhibidora del Complemento C1 , Inhibidores de Cisteína Proteinasa/farmacología , Factor XII/fisiología , Humanos , Calicreína Plasmática/antagonistas & inhibidores , Calicreína Plasmática/fisiología
9.
Thromb Haemost ; 89(5): 820-5, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12719778

RESUMEN

We have previously confirmed, using a rat mesenteric arteriole thrombolysis model, that thrombin inhibition induces endogenous thrombolysis in vivo. In addition, we have shown that thrombin-activatable fibrinolysis inhibitor (TAFI) plays a role in the down regulation of endogenous thrombolysis. However, the mechanism of endogenous thrombolysis or spontaneous plasmin generation in vivo remains unclear. It has been shown in an in vitro system that plasma kallikrein activates pro-urokinase (pro uPA) and/or plasminogen, resulting in plasmin generation. These findings suggest that spontaneous fibrinolysis might be mediated by tPA and plasma kallikrein-dependent uPA. The aim of the present study was to examine whether these mechanisms play a dominant role in endogenous thrombolysis in vivo, using our rat mesenteric arterial thrombolysis model. Argatroban infusion enhanced endogenous thrombolysis. PKSI-527, anti uPA and anti tPA IgGs suppressed argatroban-induced thrombolysis. Also, the antibody IgG preparations suppressed endogenous thrombolysis in the absence of argatroban. In the presence of PKSI-527, anti tPA IgG was more effective than anti uPA IgG in suppressing argatroban-induced thrombolysis. The results suggested that both tPA and plasma kallikrein-mediated uPA activation and tPA release contribute to endogenous fibrinolytic or thrombolytic mechanisms.


Asunto(s)
Fibrinólisis , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Ácidos Pipecólicos/farmacología , Calicreína Plasmática/fisiología , Activador de Tejido Plasminógeno/fisiología , Ácido Tranexámico/análogos & derivados , Ácido Tranexámico/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Animales , Anticuerpos/farmacología , Arginina/análogos & derivados , Antagonismo de Drogas , Fibrinolisina/biosíntesis , Fibrinólisis/efectos de los fármacos , Fibrinolíticos/farmacología , Calicreínas/antagonistas & inhibidores , Calicreínas/fisiología , Masculino , Arterias Mesentéricas , Ratas , Ratas Wistar , Sulfonamidas , Terapia Trombolítica/métodos , Trombosis , Activador de Tejido Plasminógeno/inmunología , Activador de Plasminógeno de Tipo Uroquinasa/inmunología
10.
Int Immunopharmacol ; 2(13-14): 1841-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12489798

RESUMEN

Understanding the importance and physiologic activity of the plasma kallikrein/kinin system (KKS) has been thwarted by the absence of an inclusive theory for its assembly and activation. The contact activation hypothesis describes the assembly and activation of this system in test tubes and disease states, but not under physiologic circumstances. Recent investigations have indicated a new cohesive hypothesis for understanding physiologic activation of this system. Prekallikrein (PK) and factor XI (FXI) through high molecular weight kininogen (HK) assemble on a co-localized, multiprotein receptor complex on endothelial cells that consists of at least cytokeratin 1 (CKI), gClqR, and urokinase plasminogen activator receptor (muPAR). When assembled on these proteins, prekallikrein becomes activated to kallikrein by the membrane-expressed enzyme prolylcarboxypeptidase (PRCP). Formed kallikrein then activates factor XII (FXII) for amplification of its activation and single chain urokinase. The plasma kallikrein/kinin system may serve as a physiologic counterbalance to the plasma renin angiotensin system (RAS) by lowering blood pressure and preventing thrombosis. Insights into the integrated role of these two systems may afford the development of novel therapeutic drugs to manage hypertension and thrombosis.


Asunto(s)
Endotelio/fisiología , Sistema Calicreína-Quinina/fisiología , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa , Calicreína Plasmática/metabolismo , Animales , Sitios de Unión , Unión Competitiva , Membrana Celular/metabolismo , Membrana Celular/fisiología , Endotelio/citología , Endotelio/metabolismo , Factor XI/metabolismo , Factor XI/fisiología , Factor XII/metabolismo , Factor XII/fisiología , Humanos , Sistema Calicreína-Quinina/efectos de los fármacos , Quininógeno de Alto Peso Molecular/metabolismo , Quininógeno de Alto Peso Molecular/fisiología , Calicreína Plasmática/fisiología , Precalicreína/metabolismo , Precalicreína/fisiología
11.
Immunopharmacology ; 43(2-3): 103-8, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10596840

RESUMEN

To ascertain the participation of the plasma kallikrein-kinin system (KKS) in arthritis and inflammatory bowel disease, we used two rat models resembling rheumatoid arthritis and Crohn's disease. Proteoglycan-polysaccharide from group A streptococcus (PG-APS) produced chronic destructive inflammation and systemic response in the genetically susceptible Lewis rat, in the joints when injected intraperitoneally and in the bowel when injected into the gut wall. In both models, the KKS is activated, as evidenced by decreased prekallikrein, factor XI and high molecular weight kininogen. A specific plasma kallikrein inhibitor, Bz-Pro-Phe-boroarginine, reverses the plasma changes as well as the clinical gross and microscopic pathology of both the experimental arthritis and the inflammatory bowel disease in the genetically susceptible rats. We have also shown that the tissue kallikrein system is involved in the intestinal inflammatory changes. Intestinal tissue kalikrein (ITK) is localized in goblet cells in both normal and inflamed tissue. In chronic granulomatous inflammation, ITK is localized in macrophages. ITK decreases in chronic inflammation, probably due to secretion, since the mRNA is unchanged. Kallikrein binding protein, the ITK inhibitor, decreases due to enzyme-inhibitor complexes. Both plasma and tissue kallikrein are appealing targets for drug therapy of rheumatoid arthritis and Crohn's disease.


Asunto(s)
Artritis Reumatoide/etiología , Enfermedades Inflamatorias del Intestino/etiología , Calicreína Plasmática/fisiología , Calicreínas de Tejido/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Intestinos/química , Calicreína Plasmática/análisis , Calicreína Plasmática/antagonistas & inhibidores , Polisacáridos Bacterianos/toxicidad , Proteoglicanos/toxicidad , Ratas , Ratas Endogámicas Lew , Calicreínas de Tejido/análisis , Calicreínas de Tejido/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA