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.
Pediatr Cardiol ; 35(7): 1108-15, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24714980

RESUMEN

Cardiopulmonary bypass (CPB) reduces coagulation factor levels through hemodilution and consumption. Differences in CPB-induced alterations of factor XIII (FXIII) levels in children with cyanotic and acyanotic congenital heart defects (CHDs) are not well characterized. FXIII activity (determined by Berichrom assay), prothrombin index, activated partial thromboplastin time, and fibrinogen were measured before open heart surgery with CPB and 5 days postoperatively for children older than 3 years with acyanotic (n = 30) and cyanotic (n = 30) CHDs. The preoperative FXIII levels did not differ significantly among the children of the compared groups. The cyanotic patients showed a significantly longer duration of CPB (111.4 ± 45.8 vs 71.5 ± 33.6 min; p = 0.026) and aortic cross-clamp (68.0 ± 27.1 vs 45.4 ± 31.4 min; p = 0.034). The drop in FXIII levels after termination of CPB was more profound for the children with cyanotic CHDs (87.1 ± 13.4 to 49.1 ± 13.2 vs 81.5 ± 12.6 to 58.6 ± 11.1 %, respectively; p = 0.018). The cyanotc patients also were restored to their baseline FXIII levels later than the children with acyanotic CHDs (at 48 vs 24 h). The post-CPB dynamics of the majority of the other coagulation parameters in the compared groups of patients were similar. The cyanotic patients experienced significantly greater postoperative blood loss than the acyanotic patients (12.6 ± 4.9 vs 5.0 ± 2.1 mL/kg; p < 0.001) and were transfused with larger volumes of red blood cells (10.4 ± 6.5 vs 4.2 ± 2.5 mL/kg; p = 0.007). The decrease in FXIII levels after CPB is more profound and lasts longer in children with cyanotic CHDs than in acyanotic patients. The rational strategy of postoperative FXIII replacement therapy for these categories of patients needs to be determined.


Asunto(s)
Coagulación Sanguínea/fisiología , Procedimientos Quirúrgicos Cardíacos/métodos , Cianosis/sangre , Factor XIII/metabolismo , Cardiopatías Congénitas/cirugía , Adolescente , Biomarcadores/sangre , Niño , Preescolar , Cianosis/etiología , Cianosis/cirugía , Femenino , Estudios de Seguimiento , Cardiopatías Congénitas/sangre , Cardiopatías Congénitas/complicaciones , Humanos , Masculino , Periodo Posoperatorio , Pronóstico , Estudios Prospectivos
2.
J Burn Care Res ; 35(4): 328-36, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24043240

RESUMEN

The authors have previously shown that recombinant factor XIII (rFXIII) eliminates early manifestations of multiple-organ injury caused by experimental superior mesenteric artery occlusion or trauma-hemorrhagic shock. The aim of the present study was to test the hypothesis that rFXIII provides similar protective effect in experimental burn injury. Rats were randomly divided into five groups (eight animals per group): group 1: burn + placebo treatment; group 2: burn + rFXIII pretreatment; group 3: burn + rFXIII treatment; group 4: sham burn + placebo treatment, and group 5: sham burn + rFXIII treatment. Burn (40% of TBSA) was achieved by immersing the back and abdomen of a rat into 97°C water for 10 and 5 seconds, respectively. Infusion of rFXIII (1 mg/kg) or placebo was performed immediately after burn/sham burn in treatment groups or 24 hours before burn and repeated immediately after it in pretreatment group. Endpoint parameters measured 3 hours after burn/sham burn included muscle blood flow and PO2, lung permeability, gut histology, lung and gut myeloperoxidase activity, neutrophil respiratory burst, and FXIII activity. Both treatment and pretreatment with rFXIII partially preserved microvascular blood flow in the muscle. Muscle PO2 in pretreated rats did not differ from that in shams. Pretreatment but not treatment with rFXIII preserved lung permeability. rFXIII did not have any protective effect on other endpoint parameters. In contrast to superior mesenteric artery occlusion and trauma-hemorrhagic shock experimental models, rFXIII at the doses tested has a limited effect on preventing early manifestations of multiple-organ injury after experimental burn.


Asunto(s)
Quemaduras/complicaciones , Factor XIII/farmacología , Insuficiencia Multiorgánica/prevención & control , Proteínas Recombinantes/farmacología , Daño por Reperfusión/complicaciones , Choque Hemorrágico/complicaciones , Animales , Citometría de Flujo , Íleon/metabolismo , Íleon/patología , Pulmón/metabolismo , Masculino , Microcirculación/efectos de los fármacos , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Neutrófilos/metabolismo , Oxígeno/metabolismo , Presión Parcial , Permeabilidad/efectos de los fármacos , Peroxidasa/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos
3.
Blood ; 121(11): 2108-16, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23335368

RESUMEN

Frequent infusions of intravenous factor VIII (FVIII) are required to prevent bleeding associated with hemophilia A. To reduce the treatment burden, recombinant FVIII with a longer half-life was developed without changing the protein structure. FVIII-polyethylene glycol (PEG) conjugates were prepared using an enzymatic process coupling PEG (ranging from 10 to 80 kDa) selectively to a unique O-linked glycan in the FVIII B-domain. Binding to von Willebrand factor (VWF) was maintained for all conjugates. Upon cleavage by thrombin, the B-domain and the associated PEG were released, generating activated FVIII (FVIIIa) with the same primary structure and specific activity as native FVIIIa. In both FVIII- and VWF-deficient mice, the half-life was found to increase with the size of PEG. In vivo potency and efficacy of FVIII conjugated with a 40-kDa PEG (N8-GP) and unmodified FVIII were not different. N8-GP had a longer duration of effect in FVIII-deficient mouse models, approximately a twofold prolonged half-life in mice, rabbits, and cynomolgus monkeys; however, the prolongation was less pronounced in rats. Binding capacity of N8-GP on human monocyte-derived dendritic cells was reduced compared with unmodified FVIII, resulting in several-fold reduced cellular uptake. In conclusion, N8-GP has the potential to offer efficacious prevention and treatment of bleeds in hemophilia A at reduced dosing frequency.


Asunto(s)
Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Polietilenglicoles/uso terapéutico , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Factor VIII/administración & dosificación , Factor VIII/metabolismo , Femenino , Glicosilación , Hemofilia A/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Polietilenglicoles/administración & dosificación , Polietilenglicoles/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Resultado del Tratamiento
4.
Thromb Haemost ; 101(5): 818-26, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19404533

RESUMEN

The mechanism for the elimination of factor VII (FVII) from the circulation is unknown, just as it is unclear how activation of FVII to FVIIa and subsequent complex formation with antithrombin III (AT) or alpha2-macroglobulin (alpha2M) affects clearance. The possibility that the clearance mechanism involves activation and inhibitor complex formation as obligatory intermediate reactions is examined in this study. Human and murine sera were spiked with human FVIIa in the absence and presence of heparin and analysed for complex formation. Complex formation in vivo was studied after intravenous injection of (125)I-VIIa in mice; and the pharmacokinetics (PK) of human and murine FVIIa was studied in normal mice. Furthermore, comparative PK studies were performed with FVII, FVIIa, active site blocked FVIIa and a preformed FVIIa-AT complex in normal and alpha2M-deficient mice. The data demonstrated that FVIIa-AT complexes and to a much lesser extent FVIIa-alpha2M-complexes accumulated in vivo after FVIIa administration. FVIIa-AT accounted for about 50% of total FVIIa antigen left in the circulation after 3 hours. All FVII derivatives studied including FVII, FVIIa and FVIIa-AT were cleared with similar rates suggesting an elimination kinetics which is unaffected by FVII activation and subsequent inactivation by plasma inhibitors.


Asunto(s)
Antitrombina III/metabolismo , Factor VII/farmacocinética , Factor VIIa/farmacocinética , Seroglobulinas/metabolismo , alfa-Macroglobulinas/metabolismo , Animales , Factor VIIa/administración & dosificación , Heparina/sangre , Humanos , Inyecciones Intravenosas , Radioisótopos de Yodo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Seroglobulinas/deficiencia , Seroglobulinas/genética , alfa-Macroglobulinas/deficiencia , alfa-Macroglobulinas/genética
5.
Shock ; 31(6): 621-6, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18948851

RESUMEN

Plasma factor XIII (FXIII) is responsible for stabilization of fibrin clot at the final stage of blood coagulation. Because FXIII has also been shown to modulate inflammation and endothelial permeability, we hypothesized that FXIII diminishes multiple organ dysfunction caused by gut I/R injury. A model of superior mesenteric artery occlusion (SMAO) was used to induce gut I/R injury. Rats were subjected to 45-min SMAO or sham SMAO and treated with recombinant human FXIII A2 subunit (rFXIII) or placebo at the beginning of the reperfusion period. Lung permeability, lung and gut myeloperoxidase activity, gut histology, neutrophil respiratory burst, and microvascular blood flow in the liver and muscles were measured after a 3-h reperfusion period. The effect of activated rFXIII on transendothelial resistance of human umbilical vein endothelial cells was tested in vitro. Superior mesenteric artery occlusion-induced lung permeability as well as lung and gut myeloperoxidase activity was significantly lower in rFXIII-treated versus untreated animals. Similarly, rFXIII-treated rats had lower neutrophil respiratory burst activity and ileal mucosal injury. Rats treated with rFXIII also had higher liver microvascular blood flow compared with the placebo group. Superior mesenteric artery occlusion did not cause FXIII consumption during the study period. In vitro, activated rFXIII caused a dose-dependent increase in human umbilical vein endothelial cell monolayer resistance to thrombin-induced injury. Thus, administration of rFXIII diminishes SMAO-induced multiple organ dysfunction in rats, presumably by preservation of endothelial barrier function and the limitation of polymorphonuclear leukocyte activation.


Asunto(s)
Factor XIII/farmacología , Insuficiencia Multiorgánica/tratamiento farmacológico , Insuficiencia Multiorgánica/etiología , Proteínas Recombinantes/farmacología , Daño por Reperfusión/fisiopatología , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Arteria Mesentérica Superior , Microcirculación/efectos de los fármacos , Neutrófilos/metabolismo , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos , Estallido Respiratorio/efectos de los fármacos
6.
Br J Haematol ; 143(4): 559-69, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18950467

RESUMEN

The haemostatic effect of recombinant activated factor VII (rFVIIa;NovoSeven) in thrombocytopenic patients has been a matter of controversy. Haemostasis by rFVIIa occurs via FVIIa-mediated thrombin generation in a platelet-dependent manner and may therefore be suboptimal in patients without functional platelets. Under such conditions, a clot-stabilizing agent, such as factor XIII (FXIII), may supplement the effect ofrFVIIa and improve haemostasis. Recombinant factor XIII (rFXIII-A2) is produced as an A2 homodimer of the FXIII A subunit and is equivalent to cellular FXIII normally found in platelets. The combined effects of rFVIIa andrFXIII-A2 were evaluated in clot lysis assays using factor XIII-deficient plasma and by whole blood thrombelastography (TEG) analysis from normal donors and thrombocytopenic stem cell transplantation patients. Clotting time was shortened by rFVIIa (0.6-10 microg/ml). rFVIIa only modestly improved anti-fibrinolysis,whereas rFXIII-A2 (0-20 microg/ml) enhanced anti-fibrinolysis without effect on clotting time. TEG analysis showed rFVIIa shortened the clotting time, and enhanced clot development, maximal mechanical strength and resistance to fibrinolysis, whereas, rFXIII-A2 enhanced clot development,maximal mechanical strength and markedly enhanced resistance to fibrinolysis. These data illustrate that rFVIIa and rFXIII-A2 contribute to clot formation and stability by different mechanisms suggesting enhanced haemostatic efficacy by combining these agents.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Factor VIIa/farmacología , Factor XIIIa/farmacología , Trombocitopenia/sangre , Recuento de Células Sanguíneas , Relación Dosis-Respuesta a Droga , Fibrinólisis/efectos de los fármacos , Humanos , Proteínas Recombinantes/farmacología , Trasplante de Células Madre , Tromboelastografía/métodos , Trombocitopenia/terapia
7.
Blood ; 102(13): 4393-8, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12933577

RESUMEN

Intravenous administration of recombinant human factor IX (rhFIX) acutely corrects the coagulopathy in hemophilia B dogs. To date, 20 of 20 dogs developed inhibitory antibodies to the xenoprotein, making it impossible to determine if new human FIX products, formulations, or methods of chronic administration can reduce bleeding frequency. Our goal was to determine whether hemophilia B dogs rendered tolerant to rhFIX would have reduced bleeding episodes while on sustained prophylactic rhFIX administered subcutaneously. Reproducible methods were developed for inducing tolerance to rhFIX in this strain of hemophilia B dogs, resulting in a significant reduction in the development of inhibitors relative to historical controls (5 of 12 versus 20 or 20, P <.001). The 7 of 12 tolerized hemophilia B dogs exhibited shortened whole blood clotting times (WBCTs), sustained detectable FIX antigen, undetectable Bethesda inhibitors, transient or no detectable antihuman FIX antibody titers by enzyme-linked immunosorbent assay (ELISA), and normal clearance of infused rhFIX. Tolerized hemophilia B dogs had 69% reduction in bleeding frequency in year 1 compared with nontolerized hemophilia B dogs (P =.0007). If proven safe in human clinical trials, subcutaneous rhFIX may provide an alternate approach to prophylactic therapy in selected patients with hemophilia B.


Asunto(s)
Factor IX/uso terapéutico , Hemofilia B/tratamiento farmacológico , Hemorragia/prevención & control , Animales , Animales Endogámicos , Animales Recién Nacidos , Anticuerpos Heterófilos/biosíntesis , Anticuerpos Heterófilos/inmunología , Modelos Animales de Enfermedad , Perros , Factor IX/administración & dosificación , Factor IX/inmunología , Hemofilia B/complicaciones , Hemorragia/etiología , Humanos , Tolerancia Inmunológica , Inyecciones Subcutáneas , Masculino , Proyectos Piloto , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Reproducibilidad de los Resultados
8.
Blood ; 102(2): 436-41, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12649145

RESUMEN

Recombinant human interleukin-11 (rhIL-11), a glycoprotein 130 (gp130)-signaling cytokine approved for treatment of thrombocytopenia, also raises von Willebrand factor (VWF) and factor VIII (FVIII) by an unknown mechanism. Desmopressin (1-deamino-8-d-arginine vasopressin [DDAVP]) releases stored VWF and FVIII and is used for treatment of VWF and FVIII deficiencies. To compare the effect of these 2 agents, heterozygous von Willebrand disease (VWD) and normal dogs were treated with either rhIL-11 (50 microg/kg/d subcutaneously x 7 days) or DDAVP (5 microg/kg/d intravenously x 7 days). The rhIL-11 produced a gradual and sustained elevation of VWF and FVIII levels in both heterozygous VWD and normal dogs while DDAVP produced a rapid and unsustained increase. Importantly, rhIL-11 treatment produced a 2.5- to 11-fold increase in VWF mRNA in normal canine heart, aorta, and spleen but not in homozygous VWD dogs, thus identifying a mechanism for elevation of plasma VWF in vivo. Moreover, dogs pretreated with rhIL-11 retain a DDAVP-releasable pool of VWF and FVIII, suggesting that rhIL-11 does not significantly alter trafficking of these proteins to or from storage pools. The half-life of infused VWF is unchanged by rhIL-11 in homozygous VWD dogs. These results show that rhIL-11 and DDAVP raise plasma VWF by different mechanisms. Treatment with rhIL-11 with or without DDAVP may provide an alternative to plasma-derived products for some VWD and hemophilia A patients if it is shown safe in clinical trials.


Asunto(s)
Desamino Arginina Vasopresina/uso terapéutico , Interleucina-11/uso terapéutico , ARN Mensajero/biosíntesis , Cuerpos de Weibel-Palade/metabolismo , Enfermedades de von Willebrand/fisiopatología , Factor de von Willebrand/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Desamino Arginina Vasopresina/farmacología , Perros , Evaluación Preclínica de Medicamentos , Factor VIII/metabolismo , Semivida , Corazón/efectos de los fármacos , Heterocigoto , Interleucina-11/farmacología , Miocardio/metabolismo , ARN Mensajero/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Bazo/efectos de los fármacos , Bazo/metabolismo , Cuerpos de Weibel-Palade/efectos de los fármacos , Enfermedades de von Willebrand/tratamiento farmacológico , Enfermedades de von Willebrand/genética , Factor de von Willebrand/biosíntesis , Factor de von Willebrand/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA