Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Environ Health ; 76(6): 18-27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24645409

RESUMEN

The authors conducted a survey of small streams to evaluate the effects of centralized and onsite wastewater treatment on the occurrence of selected traditional and emerging contaminants in small streams in the upper Neuse River basin, North Carolina. An undeveloped site was included to assess effects of residential land use activities on stream quality. Concentrations of nutrients and ions were higher in samples from streams in residential sites than from the stream in an undeveloped area. Overall, streams draining residential areas showed relatively small differences with respect to type of wastewater treatment. Two sites, however--one in an area of centralized wastewater treatment apparently near a suspected sewer line leak, and the second in an area of onsite wastewater treatment--showed effects of wastewater. Organic wastewater compounds were detected more frequently in samples from these two sites than from the other sites. Optical brighteners levels were correlated (r2 = .88) with the number of organic wastewater and pharmaceutical compounds detected at the residential sites and could potentially serve as a screening method to assess wastewater effects on small streams.


Asunto(s)
Ríos/química , Eliminación de Residuos Líquidos/métodos , Aguas Residuales/química , Contaminantes Químicos del Agua/análisis , North Carolina , Compuestos Orgánicos/análisis , Preparaciones Farmacéuticas/análisis , Ríos/microbiología , Aguas Residuales/análisis , Aguas Residuales/microbiología
2.
J Thromb Haemost ; 5(7): 1394-400, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17445091

RESUMEN

BACKGROUND: Activated protein C (APC) protects the host from severe sepsis. Endothelial protein C receptor (EPCR) is expressed on both hematopoietic leukocytes and non-hematopoietic endothelium, and plays a key role in protein C activation. OBJECTIVES: We explore the influence of EPCR deletion on the responses to lipopolysaccharide (LPS) and then determine whether the observed differences are due to loss of hematopoietic or non-hematopoietic EPCR. METHODS AND RESULTS: After LPS challenge, EPCR null (Procr(-/-)) mice exhibited more thrombin and cytokine generation, neutrophil sequestration in the lung and a higher mortality rate than Procr(+/-) mice. Procr(+/-) BM/Procr(-/-) (non-hematopoietic Procr(-/-)) and Procr(-/-) BM/Procr(+/-) (hematopoietic Procr(-/-)) chimeric mice were generated by bone marrow (BM) transplantation. Compared with control Procr(+/-) mice, non-hematopoietic Procr(-/-) mice exhibited reduced protein C activation by thrombin and exaggerated responses to LPS challenge, whereas Procr(+/-) mice and hematopoietic Procr(-/-) mice exhibited similar protein C activation by thrombin and similar responses to LPS challenge. CONCLUSIONS: EPCR deletion exaggerates the host responses to LPS primarily due to deficiency of EPCR on the non-hematopoietic cells.


Asunto(s)
Endotoxemia/sangre , Glicoproteínas/metabolismo , Proteína C/metabolismo , Animales , Coagulación Sanguínea , Citocinas/biosíntesis , Receptor de Proteína C Endotelial , Endotelio Vascular/metabolismo , Endotoxemia/etiología , Endotoxemia/metabolismo , Endotoxemia/patología , Glicoproteínas/deficiencia , Glicoproteínas/genética , Inflamación/sangre , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Noqueados , Receptores de Superficie Celular , Trombina/metabolismo , Trombina/farmacología
3.
J Thromb Haemost ; 3(7): 1351-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15978090

RESUMEN

Previous studies have shown that blocking endothelial protein C receptor (EPCR)-protein C interaction results in about an 88% decrease in circulating activated protein C (APC) levels generated in response to thrombin infusion and exacerbates the response to Escherichia coli. To determine whether higher levels of EPCR expression on endothelial cells might further enhance the activation of protein C and protect the host during septicemia, we generated a transgenic mouse (Tie2-EPCR) line which placed the expression of EPCR under the control of the Tie2 promoter. The mice express abundant EPCR on endothelial cells not only on large vessels, but also on capillaries where EPCR is generally low. Tie2-EPCR mice show higher levels of circulating APC after thrombin infusion. Upon infusion with factor Xa and phospholipids, Tie2-EPCR mice generate more APC, less thrombin and are protected from fibrin/ogen deposition compared with wild type controls. The Tie2-EPCR animals also generate more APC upon lipopolysaccharide (LPS) challenge and have a survival advantage. These results reveal that overexpression of EPCR can protect animals against thrombotic or septic challenge.


Asunto(s)
Factores de Coagulación Sanguínea/biosíntesis , Endotoxinas/metabolismo , Hemostasis , Receptor TIE-2/genética , Receptores de Superficie Celular/biosíntesis , Animales , Anticuerpos Monoclonales/química , Separación Celular , Progresión de la Enfermedad , Endotelio Vascular/citología , Escherichia coli/metabolismo , Fibrina/metabolismo , Fibrinógeno/metabolismo , Citometría de Flujo , Hemostáticos , Lipopolisacáridos/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteína C/metabolismo , Receptor TIE-2/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sepsis , Trombina/metabolismo , Trombosis , Factores de Tiempo , Transgenes
4.
J Thromb Haemost ; 1(4): 662-70, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12871399

RESUMEN

Activated protein C (APC) serves as an 'on demand' anticoagulant. Defects in the APC anticoagulant pathway are underlying risk factors for the development of venous and arterial thrombosis. APC has recently been shown to significantly reduce mortality in patients with severe sepsis, presumably by virtue of its ability to down-regulate coagulation as well as inflammation. Our objective was to develop an assay that, for the first time, permits rapid detection of plasma APC. This assay will expedite studies of APC in a variety of vascular disease states including sepsis, severe atherosclerosis, diabetes, and vasculitis. By generating a highly APC-specific monoclonal antibody (HAPC 1555), we have developed an assay that, for the first time, allows rapid detection of plasma APC. The Kd measured for the interaction between APC and HAPC 1555 based on BIAcore studies and binding to immobilized HAPC on microtiter plates is 6.2 +/- 0.9 and 8.8 +/- 1.0 nmol L(-1), respectively. The interaction between HAPC 1555 and APC is Ca2+-dependent, with a Ca2+ concentration of 313 +/- 48 micro mol L(-1) required for half maximal binding. HAPC 1555 interferes with APC-mediated inactivation of factor (F)Va in the presence and absence of phospholipids, suggesting that HAPC 1555 binds to the FVa binding domain of APC. When HAPC 1555 was used in an APC enzyme capture assay, therapeutic APC levels could be measured in 1.5 h, and physiologic levels of APC could be detected between 3 and 19 h. APC levels were also shown to vary markedly in patients with severe sepsis. The rapidity of our APC assay makes APC detection in patients practical clinically. This assay will expedite studies of APC in a variety of vascular disease states including sepsis, severe atherosclerosis, diabetes, and vasculitis.


Asunto(s)
Anticuerpos Monoclonales , Epítopos , Factor Va/metabolismo , Proteína C/análisis , Afinidad de Anticuerpos/efectos de los fármacos , Especificidad de Anticuerpos , Sitios de Unión , Calcio/farmacología , Estudios de Casos y Controles , Humanos , Técnicas para Inmunoenzimas/normas , Proteína C/inmunología , Proteína C/metabolismo , Sepsis/sangre , Factores de Tiempo
5.
Am J Pathol ; 159(3): 797-802, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11549570

RESUMEN

Coronary atherosclerosis with occlusive thrombosis is the major cause of acute myocardial infarction. Although plaque rupture is usually hypothesized to be the predisposing event in coronary thrombosis, the possibility cannot be excluded that local changes in the anticoagulant properties of the endothelium overlying the plaque contribute to this process. It is evident that thrombomodulin and the endothelial cell protein C receptor are critical players in the control of the thrombogenic process. This study examined whether thrombomodulin and the endothelial cell protein C receptor are down-regulated on endothelial cells overlying the atherosclerotic plaque in coronary arteries and thus could potentially favor local thrombus formation. Sections of archival left and right coronary arteries (n = 18 each) with severe atherosclerosis from the native heart of six patients who underwent heart transplantation were immunostained for CD31, CD34, endothelial cell protein C receptor, and thrombomodulin using a streptavidin-biotin-peroxidase method. Controls included left and right coronary arteries from autopsy cases with no atherosclerosis (n = 6), and also from cases with mild atherosclerosis (n = 5). The apparent density of all of these proteins was much higher in control than in atherosclerotic arteries. Our findings support the hypothesis that both endothelial cell protein C receptor and thrombomodulin are down-regulated in coronary arteries with atherosclerosis. These changes would be expected to result in reduced inhibition of thrombogenic and anti-inflammatory activity on the endothelium overlying atherosclerotic regions and thus could contribute to coronary thrombosis.


Asunto(s)
Factores de Coagulación Sanguínea , Enfermedad de la Arteria Coronaria/metabolismo , Regulación hacia Abajo , Endotelio Vascular/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de Superficie Celular/metabolismo , Trombomodulina/metabolismo , Antígenos CD34/metabolismo , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad
6.
Blood ; 97(6): 1685-8, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11238108

RESUMEN

Endothelial cell protein C receptor (EPCR) augments protein C activation by the thrombin-thrombomodulin complex about 5-fold in vitro. Augmentation is EPCR concentration dependent even when the EPCR concentration is in excess of the thrombomodulin. EPCR is expressed preferentially on large blood vessel endothelium, raising questions about the importance of protein C-EPCR interaction for augmenting systemic protein C activation. In these studies, this question was addressed directly by infusing thrombin into baboons in the presence or absence of a monoclonal antibody to EPCR that blocks protein C binding. Activated protein C levels were then measured directly by capturing the enzyme on a monoclonal antibody and assaying with chromogenic substrate. Blocking protein C-EPCR interaction resulted in about an 88% decrease in circulating activated protein C levels generated in response to thrombin infusion. Leukocyte changes, fibrinogen consumption, fibrin degradation products, and vital signs were similar between the animals infused with thrombin alone and those infused with thrombin and the anti-EPCR antibody. The results indicate that EPCR plays a major role in protein C activation and suggest that defects in the EPCR gene might contribute to increased risk of thrombosis.


Asunto(s)
Factores de Coagulación Sanguínea , Proteína C/metabolismo , Receptores de Superficie Celular/fisiología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticoagulantes/metabolismo , Bovinos , Activación Enzimática/efectos de los fármacos , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Fibrinógeno/metabolismo , Hemostáticos/administración & dosificación , Hemostáticos/farmacología , Papio , Tiempo de Tromboplastina Parcial , Proteína C/efectos de los fármacos , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Trombina/administración & dosificación , Trombina/farmacología , Trombosis/sangre , Trombosis/etiología , Factores de Tiempo
7.
J Biol Chem ; 276(11): 8364-70, 2001 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-11099506

RESUMEN

The endothelial cell protein C receptor (EPCR) is an endothelial cell-specific transmembrane protein that binds both protein C and activated protein C (APC). EPCR regulates the protein C anticoagulant pathway by binding protein C and augmenting protein C activation by the thrombin-thrombomodulin complex. EPCR is homologous to the MHC class 1/CD1 family, members of which contain two alpha-helices that sit upon an 8-stranded beta-sheet platform. In this study, we identified 10 residues that, when mutated to alanine, result in the loss of protein C/APC binding (Arg-81, Leu-82, Val-83, Glu-86, Arg-87, Phe-146, Tyr-154, Thr-157, Arg-158, and Glu-160). Glutamine substitutions at the four N-linked carbohydrate attachment sites of EPCR have little affect on APC binding, suggesting that the carbohydrate moieties of EPCR are not critical for ligand recognition. We then mapped the epitopes for four anti-human EPCR monoclonal antibodies (mAbs), two of which block EPCR/Fl-APC (APC labeled at the active site with fluorescein) interactions, whereas two do not. These epitopes were localized by generating human-mouse EPCR chimeric proteins, since the mAbs under investigation do not recognize mouse EPCR. We found that 5 of the 10 candidate residues for protein C/APC binding (Arg-81, Leu-82, Val-83, Glu-86, Arg-87) colocalize with the epitope for one of the blocking mAbs. Three-dimensional molecular modeling of EPCR indicates that the 10 protein C/APC binding candidate residues are clustered at the distal end of the two alpha-helical segments. Protein C activation studies on 293 cells that coexpress EPCR variants and thrombomodulin demonstrate that protein C binding to EPCR is necessary for the EPCR-dependent enhancement in protein activation by the thrombin-thrombomodulin complex. These studies indicate that EPCR has exploited the MHC class 1 fold for an alternative and possibly novel mode of ligand recognition. These studies are also the first to identify the protein C/APC binding region of EPCR and may provide useful information about molecular defects in EPCR that could contribute to cardiovascular disease susceptibility.


Asunto(s)
Factores de Coagulación Sanguínea , Antígenos de Histocompatibilidad Clase I/metabolismo , Proteína C/metabolismo , Receptores de Superficie Celular/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Bovinos , Glicosilación , Humanos , Ratones , Datos de Secuencia Molecular , Inhibidor de Proteína C , Receptores de Superficie Celular/metabolismo
8.
Blood ; 95(5): 1680-6, 2000 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-10688824

RESUMEN

The influence of the endothelial protein C receptor (EPCR) on the host response to Escherichia coli was studied. Animals were treated with 4 separate protocols for survival studies and analysis of physiologic and biochemical parameters: (1) monoclonal antibody (mAb) that blocks protein C/activated protein C binding to EPCR plus sublethal numbers of E coli (SLEC) (n = 4); (2) mAb to EPCR that does not block binding plus SLEC (n = 3); (3) SLEC alone (n = 4); and (4) blocking mAB alone (n = 1). Those animals receiving blocking mAb to EPCR plus sublethal E coli died 7 to 54 hours after challenge, whereas all animals treated with the other protocols were permanent survivors. Histopathologic studies of tissues from animals receiving blocking mAb plus SLEC removed at postmortem were compared with those animals receiving SLEC alone killed at T+24 hours. The animals receiving the blocking mAb exhibited consumption of fibrinogen, microvascular thrombosis with hemorrhage of both the adrenal and renal cortex, and an intense influx of neutrophils into the adrenal, renal, and hepatic microvasculature, whereas the tissues from animals receiving only sublethal E coli exhibited none of these abnormal histopathologic changes. Compared with the control animals, the animals receiving the blocking mAb exhibited significantly elevated serum glutamic pyruvic transaminase, anion gap, thrombin-antithrombin complex, IL-6, IL-8, and soluble thrombomodulin. The levels of circulating activated protein C varied too widely to allow a clear determination of whether the extent of protein C activation was altered in vivo by blocking protein C binding to EPCR. We conclude that protein C/activated protein C binding to EPCR contributes to the negative regulation of the coagulopathic and inflammatory response to E coli and that EPCR provides an additional critical step in the host defense against E coli. (Blood. 2000;95:1680-1686)


Asunto(s)
Factores de Coagulación Sanguínea , Infecciones por Escherichia coli/fisiopatología , Receptores de Superficie Celular/fisiología , Sepsis/fisiopatología , Animales , Anticuerpos Monoclonales/farmacología , Antitrombina III/análisis , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/patología , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/patología , Interleucinas/sangre , Elastasa de Leucocito/sangre , Pulmón/patología , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/patología , Papio , Péptido Hidrolasas/análisis , Receptores de Superficie Celular/inmunología , Sepsis/sangre , Sepsis/microbiología , Sepsis/patología , Trombomodulina/sangre , Activador de Tejido Plasminógeno/análisis , Factor de Necrosis Tumoral alfa/análisis , Vísceras/patología , alfa 1-Antitripsina/análisis
9.
Blood ; 95(5): 1687-93, 2000 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-10688825

RESUMEN

The endothelial cell protein C receptor (EPCR) facilitates protein C activation by the thrombin-thrombomodulin complex. Protein C activation has been shown to be critical to the host defense against septic shock. In cell culture, tumor necrosis factor-alpha (TNF-alpha) down-regulates EPCR expression, raising the possibility that EPCR might be down-regulated in septic shock. We examined EPCR mRNA and soluble EPCR levels in mice and rats challenged with lethal dose 95 levels of endotoxin. Toxic doses of TNF-alpha failed to alter EPCR mRNA levels in mice. Rather than EPCR mRNA levels falling in response to endotoxin, as predicted from cell-culture experiments, they rose approximately 3-fold 6 hours after exposure to endotoxin before returning toward baseline levels at 24 hours after exposure. Soluble EPCR levels rose approximately 4-fold. Infusion of hirudin, a specific thrombin inhibitor, before endotoxin exposure almost completely blocked the increase in EPCR mRNA and soluble EPCR. Consistent with the idea that the responses were mediated by thrombin, thrombin infusion (5 U/kg of body weight for 3 hours) resulted in an approximately 2-fold increase in EPCR mRNA and soluble EPCR. Incubation of rat endothelial cells with thrombin or murine protease-activated receptor 1 agonist peptide resulted in a 2-fold increase in EPCR mRNA. These results indicate that thrombin plays a major role in up-regulating EPCR mRNA and shedding in vivo. (Blood. 2000;95:1687-1693)


Asunto(s)
Factores de Coagulación Sanguínea , Endotelio Vascular/efectos de los fármacos , Endotoxemia/metabolismo , Endotoxinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , ARN Mensajero/biosíntesis , Receptores de Superficie Celular/biosíntesis , Trombina/farmacología , Animales , Antitrombina III/metabolismo , Células CHO , Cricetinae , Cricetulus , Regulación hacia Abajo/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotoxemia/genética , Fibrinógeno/análisis , Hirudinas/farmacología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Miocardio/metabolismo , Oligopéptidos/farmacología , Péptido Hidrolasas/metabolismo , Proteína C/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-1 , Receptor PAR-2 , Receptores de Superficie Celular/genética , Receptores de Trombina/agonistas , Trombina/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
11.
Circulation ; 96(10): 3633-40, 1997 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-9396465

RESUMEN

BACKGROUND: The protein C anticoagulant pathway is critical to the control of hemostasis. Thrombomodulin and a newly identified receptor for protein C/activated protein C, EPCR, are both present on endothelium. EPCR augments activation of protein C by the thrombin-thrombomodulin complex. METHODS AND RESULTS: To gain a better understanding of the relationship between thrombomodulin and EPCR, we compared the cellular specificity and tissue distributions of these two receptors by using immunohistochemistry. EPCR expression was detected almost exclusively on endothelium in human and baboon tissues. In most organs, EPCR was expressed relatively intensely on the endothelium of all arteries and veins, most arterioles, and some postcapillary venules. EPCR staining was usually negative on capillary endothelial cells. In contrast, thrombomodulin was detected at high concentrations in both large vessels and capillary endothelium. Both thrombomodulin and EPCR were expressed poorly on brain capillaries. The liver sinusoids were the only capillaries in which EPCR was expressed at moderate levels and thrombomodulin was low. EPCR and thrombomodulin were both expressed on the endothelium of vasa recta in the renal medulla, the lymph node subcapsular and medullary sinuses, and some capillaries within the adrenal gland. Even in these organs the majority of capillaries were EPCR negative or stained weakly. CONCLUSIONS: These studies suggest that EPCR may be important in enhancing protein C activation on large vessels. The presence of high levels of EPCR on arterial vessels may help explain why partial protein C deficiency is a weak risk factor for arterial thrombosis.


Asunto(s)
Anticoagulantes/metabolismo , Endotelio Vascular/metabolismo , Proteína C/metabolismo , Receptores de Superficie Celular/metabolismo , Adulto , Animales , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Concentración Osmolar , Papio , Trombomodulina/metabolismo , Distribución Tisular
13.
Proc Natl Acad Sci U S A ; 93(19): 10212-6, 1996 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-8816778

RESUMEN

Protein C activation on the surface of the endothelium is critical to the negative regulation of blood coagulation. We now demonstrate that monoclonal antibodies that block protein C binding to the endothelial cell protein C receptor (EPCR) reduce protein C activation rates by the thrombin-thrombomodulin complex on endothelium, but that antibodies that bind to EPCR without blocking protein C binding have no effect. The kinetic result of blocking the EPCR-protein C interaction is an increased apparent Km for the activation without altering the affinity of thrombin for thrombomodulin. Activation rates of the protein C derivative lacking the gamma-carboxyglutamic acid domain, which is required for binding to EPCR, are not altered by the anti-EPCR antibodies. These data indicate that the protein C activation complex involves protein C, thrombin, thrombomodulin, and EPCR. These observations open new questions about the control of coagulation reactions on vascular endothelium.


Asunto(s)
Factores de Coagulación Sanguínea , Proteína C/metabolismo , Receptores de Superficie Celular/fisiología , Trombina/metabolismo , Trombomodulina/metabolismo , Anticuerpos Monoclonales , Sitios de Unión , Línea Celular , Endotelio Vascular/fisiología , Humanos , Cinética , Modelos Biológicos , Proteína C/farmacología , Receptores de Superficie Celular/biosíntesis , Proteínas Recombinantes/metabolismo , Transfección
14.
Thromb Haemost ; 75(4): 578-84, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8743182

RESUMEN

Inflammation often is considered a contributing factor to both thrombosis and disseminated intravascular coagulation. The molecular mechanisms that dictate which of these clinical manifestations will result from the inflammatory stimulus remain obscure. Bacterial infection and certain tumors are common initiators of the disseminated intravascular coagulant response. Complement activation resulting from bacterial infection shares with selected tumors the capacity to generate or release membrane particles that lack functional adhesion receptors and hence could circulate to amplify a disseminated intravascular coagulant response. We developed a model of venous thrombosis that resulted in localized thrombus formation without disseminated intravascular coagulation. The model involves infusion of tumor necrosis factor, blockade of protein C and a partial decrease in venous flow caused by ligation of the superficial femoral vein without obstruction of the deep formal vein. Infusion of phospholipid vesicles into this model resulted in amplification of a localized thrombotic response into a consumptive response. Seven different groups of animals were studied. The first three groups established the conditions necessary to produce deep vein thrombosis. The second four groups established the conditions necessary to produce disseminated intravascular coagulation. The infusion of phospholipid vesicles plus tumor necrosis factor and anti-protein C antibody resulted in consumption of fibrinogen, the production of thrombin/antithrombin complexes, a fall in platelet count, and venous thrombosis. Without ligation and catheterization phospholipid vesicles failed to produce the consumptive response. We conclude, therefore, that phospholipid vesicles can amplify a local thrombotic response into a consumptive response, and that vesiculation accompanying inflammation is one means by which localized coagulant activity may be amplified to produce disseminated intravascular coagulation.


Asunto(s)
Fibrinógeno/metabolismo , Fosfolípidos/toxicidad , Proteína C/inmunología , Tromboflebitis/inducido químicamente , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Anticuerpos Monoclonales , Antitrombina III/biosíntesis , Sinergismo Farmacológico , Infusiones Intravenosas , Liposomas , Papio , Péptido Hidrolasas/biosíntesis , Recuento de Plaquetas/efectos de los fármacos
15.
J Biol Chem ; 269(10): 7514-9, 1994 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-8125971

RESUMEN

The naturally occurring nullHong Kong variant of human alpha 1-antitrypsin is truncated at its carboxyl terminus, and is retained and degraded in a pre-Golgi compartment of stably transfected murine hepatoma cells (Le, A., Graham, K. S., and Sifers, R. N. (1990) J. Biol. Chem. 265, 14001-14007). Long-term metabolic radiolabeling with [35S]methionine or [32P]orthophosphate in combination with low stringency immunoprecipitation of the nullHong Kong variant has resulted in the co-precipitation of a radiolabeled 90-kDa protein designated p90. Several criteria, including mobility in SDS-polyacrylamide gel electrophoresis, absence of asparagine-linked oligosaccharides, and immunoreactivity with peptide-specific antiserum, have indicated that co-precipitating p90 is identical to calnexin, a calcium-binding phosphoprotein of the endoplasmic reticulum membrane (Wada, I. W., Rindress, P. H., Ou, W.-J., Doherty, J. J., Louvard, D., Bell, A. W., Dignard, D., Thomas, D. Y., and Bergeron, J. J. M. (1991) J. Biol. Chem. 266, 19599-19610). Finally, results from co-immunoprecipitation analyses and velocity sedimentation experiments have verified that approximately 30% of the retained nullHong Kong variant polypeptides are associated with calnexin in a 1:1 molar ratio and can be dissociated with either deoxycholate or chelation of calcium ions at 37 degrees C. Overall, these findings may extend our current understanding of the molecular pathogenesis of serum alpha 1-antitrypsin deficiency.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , alfa 1-Antitripsina/metabolismo , Animales , Calcio , Proteínas de Unión al Calcio/inmunología , Calnexina , Quelantes , Glicosilación , Humanos , Sueros Inmunes , Proteínas de la Membrana/metabolismo , Ratones , Péptidos/metabolismo , Fosforilación , Pruebas de Precipitina , Células Tumorales Cultivadas , alfa 1-Antitripsina/genética
16.
J Biol Chem ; 267(2): 1072-80, 1992 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-1530934

RESUMEN

Greater than 85% of the transport-impaired PiZ variant of human alpha 1-antitrypsin is retained within transfected mouse hepatoma cells and is subjected to intracellular degradation (Le, A., Graham, K., and Sifers, R.N. (1990) J. Biol. Chem. 265, 14001-14007). The retained protein undergoes a discrete size reduction that results from the modification of its endoglycosidase H-sensitive oligosaccharides and is inhibited by 1-deoxymannojirimycin. Metabolic poisons and inhibitors of protein synthesis perturb the intracellular degradation of the retained protein but do not affect its size reduction. The ability of metabolic poisons to influence the degradation of the PiZ variant in cells treated with brefeldin A indicates that export of the macromolecule from the endoplasmic reticulum (ER) is not the energy-dependent component of its degradation. Subcellular fractionation experiments have verified that both the size reduction and degradation of the retained PiZ variant occur within the rough ER. Finally, sedimentation velocity centrifugation analysis of radiolabeled cell extracts has indicated that approximately 80% of the PiZ variant consists as soluble aggregates immediately after its synthesis. An inability to detect more extensive aggregation during the retention period supports our previous conclusion that only a small fraction of the macromolecules actually form large insoluble aggregates (Graham, K.S., Le, A., and Sifers, R.N. (1990) J. Biol. Chem. 265, 20463-20468). Overall, these findings indicate that soluble aggregates of the PiZ variant are degraded within the ER by a mechanism sensitive to inhibitors of protein synthesis.


Asunto(s)
Retículo Endoplásmico/metabolismo , Inhibidores de la Síntesis de la Proteína , alfa 1-Antitripsina/metabolismo , 1-Desoxinojirimicina , 2,4-Dinitrofenol , Animales , Brefeldino A , Cicloheximida/farmacología , Ciclopentanos/farmacología , Dinitrofenoles/farmacología , Electroforesis en Gel de Poliacrilamida , Retículo Endoplásmico/efectos de los fármacos , Glucosamina/análogos & derivados , Glucosamina/farmacología , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/enzimología , Humanos , Neoplasias Hepáticas Experimentales/enzimología , Ratones , Nocodazol/farmacología , Puromicina/farmacología , Transfección , Células Tumorales Cultivadas , alfa 1-Antitripsina/genética
17.
Blood ; 78(2): 357-63, 1991 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-1829967

RESUMEN

Activated protein C is a plasma anticoagulant. For activated protein C to function as an anticoagulant, it must form a complex with protein S. Protein S anticoagulant activity is neutralized by formation of a reversible complex with C4b binding protein (C4bBP). C4bBP is an acute-phase plasma protein. When C4bBP levels increase, mass action forces the level of free protein S to decrease, giving rise to an acquired functional protein S deficiency. It has been proposed that these elevated C4bBP levels and the resultant acquired deficiency of protein S that occurs in inflammation could contribute to a hypercoagulable state. An experimental model to test this hypothesis was suggested by our previous studies that demonstrated that inhibition of protein C activation rendered baboons hypercoagulable in response to sublethal Escherichia coli infusion (J Clin Invest 79:918, 1987). We have extended these studies to examine the effect of inhibition of protein S activity with C4bBP in the host (baboon) response to infusion of sublethal concentrations of E coli organisms. Five sets of animals were studied: (1) those challenged with sublethal concentrations of E coli alone (0.4 x 10(10)/kg); (2) those supplemented only with C4bBP (20 mg/kg); (3) those challenged with the same level of E coli but supplemented with C4bBP (20 mg/kg); (4) those challenged with sublethal E coli and supplemented with C4bBP (20 mg/kg) and sufficient protein S (2.3 mg/kg) to fill the protein S binding sites on C4bBP; and (5) those challenged with lethal concentrations of E coli. Sublethal E coli infusion (group 1 animals) caused only an acute-phase response with no consumption of fibrinogen, detectable organ damage, or detectable tumor necrosis factor (TNF) in the plasma. C4bBP infusion (group 2 animals) resulted in no significant physiologic changes, no detectable plasma TNF, and little change in fibrinogen level. The group 3 animals, receiving both sublethal E coli and C4bBP, exhibited rapid consumption of fibrinogen, systemic organ damage, and detectable circulating TNF ultimately leading to death. The overall response of this group was very similar to the response of the group 5 animals receiving an LD100 dose of E coli. The group 4 animals, which were treated exactly as above except that C4bBP was supplemented with a slight excess of protein S, responded essentially like those that received sublethal E coli alone. These studies suggest that the elevation of C4bBP during an inflammatory response can contribute to fibrinogen consumption and vascular damage. This vascular damage may be associated with enhanced elaboration of cytokines like TNF.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Proteínas Portadoras/fisiología , Complemento C4b/fisiología , Proteínas Inactivadoras de Complemento , Infecciones por Escherichia coli/fisiopatología , Animales , Presión Sanguínea , Proteínas Portadoras/toxicidad , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/patología , Fibrinógeno/análisis , Glicoproteínas/uso terapéutico , Frecuencia Cardíaca , Humanos , Riñón/patología , Necrosis , Papio , Proteína S , Circulación Renal , Factor de Necrosis Tumoral alfa/análisis
18.
Dev Biol Stand ; 67: 51-7, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3301468

RESUMEN

Protein C and protein S serve as natural anticoagulants. Deficiencies of these proteins are often associated with recurrent deep vein thrombosis and coumarin induced skin necrosis. These two proteins function by selectively inactivating factors Va and VIIIa, two of the "cofactors" of blood coagulation. Hence, inhibition of coagulation by this pathway complements the better known inhibition mediated by the antithrombin III-heparin system. These observations suggest that protein C and/or activated protein C may prove useful in controlling thrombosis and/or DIC. We have developed a Ca2+ dependent monoclonal antibody which allows the rapid isolation of human protein C. This rapid isolation has allowed us to demonstrate that activated protein C can protect baboons from the lethal effects of E. coli/endotoxin and that protein C supplementation can minimize fibrinogen consumption following tissue factor infusion into dogs.


Asunto(s)
Proteína C/aislamiento & purificación , Tromboflebitis/prevención & control , Animales , Bovinos , Infecciones por Escherichia coli/complicaciones , Fibrinógeno/fisiología , Papio , Proteína C/uso terapéutico , Tromboflebitis/etiología
19.
J Clin Invest ; 70(1): 127-34, 1982 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-6177716

RESUMEN

An endothelial cell-associated cofactor that greatly enhances the rate of protein C activation by thrombin has recently been described. The observation that the cofactor binds thrombin with unusually high affinity (K(d) = 0.5 nM) suggested that low level thrombin infusion into dogs might lead to the selective activation of protein C. Infusion of thrombin (1 U/min per kg body wt) into the jugular vein of dogs leads to the formation of a systemic anticoagulant activity within 5 min of starting the infusion. The plasma has a prolonged partial thromboplastin time and Factor X(a) clotting time, but there is no change in the thrombin clotting time. The systemic anticoagulant activity is identified as activated protein C for the following reasons: (a) anti-canine activated protein C IgG antibodies inhibit the anticoagulant activity; (b) the anticoagulant activity can be partially purified from the plasma of dogs infused with thrombin by barium citrate adsorption; (c) the anticoagulant has chromatographic properties on QAE Sephadex indistinguishable from those of activated protein C, and (d) the rate at which this anticoagulant is inhibited in citrated canine plasma is identical to that of canine activated protein C. The in vivo activation of protein C appears to be receptor mediated since it occurs at low thrombin concentration and since it can be progressively inhibited by simultaneous infusion of diisopropylphospho-thrombin with thrombin. The activation of protein C at low levels of thrombin is selective, since neither the platelet count nor the Factor V levels are altered. Thrombin infusion leads to an elevation in circulating plasminogen activator levels. This appears to be mediated through the activation of protein C since coinfusion of diisopropylphospho-thrombin with thrombin inhibits the increase in plasminogen activator levels. Pretreatment of dogs with dicumarol blocks both the formation of anticoagulant activity and the rise in plasminogen activator. When the dicumarol-treated dogs are supplemented with isolated protein C and thrombin is infused, the anticoagulant activity again appears and the circulating levels of plasminogen activator are again elevated. These studies illustrate that low levels of thrombin in vivo can activate protein C, which in turn can inhibit blood coagulation and initiate fibrinolysis by elevating circulating plasminogen activator levels.


Asunto(s)
Glicoproteínas/biosíntesis , Animales , Coagulación Sanguínea/efectos de los fármacos , Pruebas de Coagulación Sanguínea , Bovinos , Dicumarol/farmacología , Perros , Factor V/análisis , Factor X , Factor Xa , Glicoproteínas/metabolismo , Isoflurofato/farmacología , Activadores Plasminogénicos/análisis , Activadores Plasminogénicos/biosíntesis , Recuento de Plaquetas , Proteína C , Trombina/administración & dosificación , Trombina/farmacología , Tiempo de Trombina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA