Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
N Engl J Med ; 361(4): 345-57, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19625716

RESUMEN

BACKGROUND: The hemolytic-uremic syndrome consists of the triad of microangiopathic hemolytic anemia, thrombocytopenia, and renal failure. The common form of the syndrome is triggered by infection with Shiga toxin-producing bacteria and has a favorable outcome. The less common form of the syndrome, called atypical hemolytic-uremic syndrome, accounts for about 10% of cases, and patients with this form of the syndrome have a poor prognosis. Approximately half of the patients with atypical hemolytic-uremic syndrome have mutations in genes that regulate the complement system. Genetic factors in the remaining cases are unknown. We studied the role of thrombomodulin, an endothelial glycoprotein with anticoagulant, antiinflammatory, and cytoprotective properties, in atypical hemolytic-uremic syndrome. METHODS: We sequenced the entire thrombomodulin gene (THBD) in 152 patients with atypical hemolytic-uremic syndrome and in 380 controls. Using purified proteins and cell-expression systems, we investigated whether thrombomodulin regulates the complement system, and we characterized the mechanisms. We evaluated the effects of thrombomodulin missense mutations associated with atypical hemolytic-uremic syndrome on complement activation by expressing thrombomodulin variants in cultured cells. RESULTS: Of 152 patients with atypical hemolytic-uremic syndrome, 7 unrelated patients had six different heterozygous missense THBD mutations. In vitro, thrombomodulin binds to C3b and factor H (CFH) and negatively regulates complement by accelerating factor I-mediated inactivation of C3b in the presence of cofactors, CFH or C4b binding protein. By promoting activation of the plasma procarboxypeptidase B, thrombomodulin also accelerates the inactivation of anaphylatoxins C3a and C5a. Cultured cells expressing thrombomodulin variants associated with atypical hemolytic-uremic syndrome had diminished capacity to inactivate C3b and to activate procarboxypeptidase B and were thus less protected from activated complement. CONCLUSIONS: Mutations that impair the function of thrombomodulin occur in about 5% of patients with atypical hemolytic-uremic syndrome.


Asunto(s)
Activación de Complemento/genética , Síndrome Hemolítico-Urémico/genética , Mutación Missense , Trombomodulina/genética , Adolescente , Adulto , Niño , Complemento C3b , Factor I de Complemento , Vía Alternativa del Complemento/fisiología , Análisis Mutacional de ADN , Síndrome Hemolítico-Urémico/inmunología , Heterocigoto , Humanos , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple , Trombomodulina/metabolismo , Adulto Joven
2.
Arterioscler Thromb Vasc Biol ; 29(7): 1087-92, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19372456

RESUMEN

OBJECTIVE: In spite of major advances in reperfusion therapy for patients presenting with acute coronary syndrome, long-term morbidity is still substantial. A limitation of initial treatment of myocardial ischemia is the lack of prevention of ischemia/reperfusion (I/R) injury. Activated protein C (APC), a crucial mediator in the coagulation process, plays a prominent role in the crosstalk between coagulation and inflammation and provides cytoprotective effects via inhibition of apoptosis and inflammation in several human and animal studies. METHODS AND RESULTS: APC was administered in an animal model for myocardial I/R. APC largely inhibited early myocardial I/R injury after varying reperfusion times, an effect that was absent on administration of heparin, a nonspecific anticoagulant agent. The protective effects of APC were absent in case of absence or blockade of protease activated receptor-1 (PAR-1), indicating a critical role for PAR-1 in this process. Furthermore, we showed a strong antiapoptotic effect of APC in the early phase of reperfusion combined with an antiinflammatory effect at an early stage (IL-6), as well as at a later stage (leukocyte infiltration). CONCLUSIONS: APC exerts strong protective effects on early myocardial I/R injury, primarily via inhibition of apoptosis and inflammation, which are regulated via PAR-1.


Asunto(s)
Anticoagulantes/administración & dosificación , Apoptosis/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Proteína C/administración & dosificación , Animales , Apoptosis/inmunología , Regulación de la Expresión Génica , Inflamación/fisiopatología , Inflamación/prevención & control , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/fisiopatología , Receptor PAR-1/efectos de los fármacos , Receptor PAR-1/fisiología
3.
Blood ; 109(3): 1003-9, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17023579

RESUMEN

Recent studies have shown that endothelial protein C receptor (EPCR) polymorphisms and soluble EPCR levels are associated with thrombotic diseases. It is unknown whether membrane EPCR (mEPCR) heterozygosity and/or physiologically elevated sEPCR levels directly impact the hemostatic balance and the outcome of endotoxemia. In these studies, thrombin infusion experiments revealed that EPCR heterozygosity (Procr+/-) impaired protein C activation by approximately 30%. Infusion of factor Xa with phospholipid demonstrated that the Procr+/- genotype increased the coagulant response relative to wild-type mice. Challenge of the Procr+/- mice with lipopolysaccharide (LPS) did not significantly exaggerate their response compared with wild-type mice. We also generated mice in which one allele of full-length EPCR was replaced by sEPCR (Procrs/+). Compared with Procr+/- mice, Procrs/+ mice had 5-fold higher sEPCR and similar mEPCR levels. Procr+/- and Procrs/+ mice generated similar levels of activated protein C (APC) upon thrombin infusion. They also exhibited a similar coagulant response upon factor Xa/phospholipid infusion. Only supraphysiologic levels of sEPCR could influence protein C activation and exaggerate the coagulant response. In conclusion, mEPCR, but not physiologically elevated sEPCR, regulated protein C activation. Procr heterozygosity results in a mild increase of thrombosis tendency and little influence on the response to endotoxin.


Asunto(s)
Endotoxemia/etiología , Glicoproteínas/fisiología , Hemostasis , Animales , Receptor de Proteína C Endotelial , Heterocigoto , Lipopolisacáridos/farmacología , Proteínas de la Membrana , Ratones , Ratones Mutantes , Proteína C/metabolismo , Receptores de Superficie Celular , Solubilidad , Trombina/farmacología , Trombosis/etiología
4.
Blood ; 106(8): 2716-22, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15956290

RESUMEN

The endothelial cell protein C receptor (EPCR) augments protein C activation by the thrombin-thrombomodulin complex. Deletion of the EPCR gene (Procr) in mice leads to embryonic lethality before embryonic day 10 (E10.0). EPCR is detected in the giant trophoblast cells at the feto-maternal boundary from E7.5 and weakly in embryonic aortic endothelial cells from E13.5, suggesting that extraembryonic EPCR expression may be essential for embryonic viability. Using conditional knock-out strategies, we demonstrate that Procr-deficient embryos with EPCR expression on placenta giant trophoblasts can be carried to term and then develop normally. Conversely, EPCR expression in the embryo, without expression in the giant trophoblast cells, does not rescue the mice. In genetically modified mice with low tissue factor activity, Procr deficiency is not lethal to the embryo. As adults, Procr-deficient mice generate more thrombin and activate less protein C in response to procoagulant stimuli. Spontaneous thrombin formation in the deficient animals increases with age. These findings show that extraembryonic EPCR expression is critical for embryo development.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Receptores de Superficie Celular/metabolismo , Envejecimiento/fisiología , Animales , Factores de Coagulación Sanguínea/genética , Pérdida del Embrión/genética , Embrión de Mamíferos/metabolismo , Genotipo , Ratones , Ratones Noqueados , Especificidad de Órganos , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Tromboplastina/deficiencia , Tromboplastina/genética , Trofoblastos/metabolismo
5.
Blood ; 104(13): 3958-64, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15319291

RESUMEN

Activated protein C (APC) supplementation significantly reduces mortality in patients with severe sepsis, presumably by down-regulating coagulation, inflammation, and apoptosis. In vivo, endogenous APC is generated from protein C (PC) "on demand" in response to elevated thrombin levels. Thrombomodulin and endothelial cell protein C receptor are endothelial receptors required to generate APC endogenously. Since these receptors may be down-regulated in sepsis, we measured plasma markers of APC generation in 32 patients with severe sepsis to determine whether APC generation is impaired and whether markers of APC generation correlate with 28-day mortality. Relative to normals, all patients had elevated F1 + 2 and thrombin-antithrombin complex (TAT) levels (markers of thrombin generation and inhibition, respectively), and 28 of 32 patients had reduced PC levels. In 20 patients, APC levels paralleled elevated F1 + 2 levels, whereas 12 patients had low APC levels despite elevated F1 + 2 levels, suggesting that APC generation is impaired in the latter. No significant differences exist between survivors and nonsurvivors with respect to baseline PC levels, F1 + 2 levels, and APACHE II (acute physiology and chronic health evaluation) scores. Baseline APC levels were higher in survivors (P = .024), and baseline F1 + 2/APC ratios were lower in survivors (P = .047). Larger studies are warranted to establish whether APC generation profiles aid in managing sepsis.


Asunto(s)
Proteína C/metabolismo , Sepsis/sangre , APACHE , Adulto , Anciano , Anciano de 80 o más Años , Activación Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína C/uso terapéutico , Sepsis/tratamiento farmacológico , Sepsis/mortalidad , Análisis de Supervivencia , alfa 1-Antitripsina/metabolismo
6.
J Acoust Soc Am ; 112(3 Pt 1): 1196-201, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12243164

RESUMEN

Cavitation is an effective physical mechanism for concentrating mechanical energy. Accordingly, it has wide applications in such diverse fields as sonochemistry, in which chemical reactions are initiated or accelerated, or in the electronic component cleaning industry in which particles (and other materials) are removed from surfaces. However, devices designed to act as cavitation monitors have had little success, partly because their intrusiveness often affects the cavitation field itself. Presented here is a brief description of a unique cavitation monitor that utilizes the phenomenon of sonoluminescence as an indirect quantifier of cavitation. It appears to work efficiently over a broad range of acoustic field intensities and its application to megasonic cleaning has provided interesting and valuable insights into this technology.

7.
J Biol Chem ; 277(45): 43335-43, 2002 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-12218060

RESUMEN

The endothelial cell protein C receptor (EPCR) is a type 1 transmembrane protein found primarily on endothelium that binds both protein C and activated protein C with similar affinity. EPCR augments the activation of protein C by the thrombin-thrombomodulin complex. To determine the physiological importance of EPCR, we generated EPCR-deficient mice by homologous targeting in embryonic stem cells. Genotyping of progeny obtained from EPCR(+/-) interbreeding indicated that EPCR(-/-) embryos died on or before embryonic day 10.5 (E10.5). Reverse transcriptase-PCR confirmed the absence of EPCR mRNA in EPCR(-/-) embryos. EPCR(-/-) embryos removed from extra-embryonic membranes and tissues at day E7.5 and cultured in vitro developed beyond E10.5, suggesting a role for EPCR in the normal function of the placenta and/or at the materno-embryonic interface. Immunohistochemistry revealed the lack of EPCR in trophoblast giant cells of EPCR(-/-) embryos. These cells, which normally express EPCR, are in direct contact with the maternal circulation and its clotting factors. In EPCR(-/-) embryos, greatly increased fibrin deposition was detected around these cells. To prevent this fibrin deposition, EPCR(+/-)-crossed female mice received a daily subcutaneous injection of enoxaparin through pregnancy. Although some EPCR(-/-) embryos were rescued from midgestational lethality, this regimen yielded no EPCR(-/-) pups. We conclude that EPCR is essential for normal embryonic development. Moreover, EPCR plays a key role in preventing thrombosis at the maternal-embryonic interface.


Asunto(s)
Factores de Coagulación Sanguínea/genética , Muerte Fetal , Enfermedades Placentarias/genética , Receptores de Superficie Celular/genética , Animales , Secuencia de Bases , Factores de Coagulación Sanguínea/metabolismo , Cartilla de ADN , Embrión de Mamíferos/fisiología , Femenino , Genotipo , Edad Gestacional , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Enfermedades Placentarias/patología , Embarazo , ARN Mensajero/genética , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Trombosis
8.
Thromb Haemost ; 88(2): 259-66, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12195698

RESUMEN

The endothelial cell protein C receptor (EPCR) augments protein C activation by the thrombomodulin.thrombin complex. Deletion of the EPCR gene in mice has been reported to lead to embryonic lethality before embryonic day 10 (E10.0). To identify potential mechanisms responsible for this lethality, we performed an immunohistological analysis of EPCR distribution during mouse embryogenesis. EPCR was detected in the trophoblast giant cells at the feto-maternal boundary from E7.5 and at later time points in the trophoblasts of the placenta, suggesting a role in the haemostatic regulation of the maternal blood that irrigates these surfaces. In the embryo, EPCR was weakly detected in aortic endothelial cells from E13.5. Thereafter, EPCR levels increased in certain large blood vessels endothelial cells suggesting that the specificity of EPCR to large vessels is conferred in utero. However, not until postnatal day 7 did the intensity and distribution of EPCR staining mimic that observed in adult mice.


Asunto(s)
Endotelio Vascular/química , Endotelio Vascular/embriología , Animales , Aorta/citología , Aorta/embriología , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Embrión de Mamíferos/irrigación sanguínea , Endotelio Vascular/citología , Inmunohistoquímica , Ratones , Placenta/citología , Distribución Tisular , Trofoblastos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA