Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Haemophilia ; 23(2): 292-299, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27862687

RESUMEN

INTRODUCTION: The development of neutralizing antibodies (inhibitors) against coagulation factor VIII (FVIII) is currently the most serious complication for patients with haemophilia A undergoing FVIII replacement therapy. Several genetic factors have been acknowledged as risk factors for inhibitor development. AIM: To analyze the influence of genetic factors on the nature of the humoral immune response to FVIII in eight brother pairs with inhibitors. METHODS: The domain specificity of FVIII-specific IgG was analysed by antibody binding to FVIII fragments and homologue-scanning mutagenesis (HSM). The FVIII-specific IgG subclasses were measured by direct ELISA. RESULTS: Of the 16 patient analysed with both methods, 12 had A2- and 13 had C2-specific IgG. The presence of A1-, A3- or C1-specific IgG was identified in nine of 14 patients analysed by HSM. IgG1, IgG2 and IgG4 subclasses contributed to the anti-FVIII IgG response, and the amount of FVIII-specific IgG1 (r = 0.66) and IgG4 (r = 0.69) correlated significantly with inhibitor titres. Patients with high concentrations of total anti-FVIII IgG (r = 0.69) or high inhibitor titres (r = 0.52) had a high proportion of FVIII-specific IgG4. Statistical analysis revealed trends/evidence that the subclass distribution (P = 0.0847) and domain specificity to HC/LC (P = 0.0883) and A2/C2 (P = 0.0011) of anti-FVIII IgG were more similar in brothers compared to unrelated subjects. CONCLUSION: Overall, our data provide a first hint that anti-FVIII IgG characteristics are comparable among haemophilic brothers with inhibitors. Whether genetic factors also influence the nature of patients' antibodies needs to be confirmed in a larger study population.


Asunto(s)
Anticuerpos/sangre , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Factor VIII/administración & dosificación , Hemofilia A/inmunología , Humanos , Masculino , Hermanos
2.
Haemophilia ; 20(5): 723-31, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24750465

RESUMEN

Factor VIII (FVIII) is a multidomain blood plasma glycoprotein. Activated FVIII acts as a cofactor to the serine protease factor IXa within the membrane-bound tenase complex assembled on the activated platelet surface. Defect or deficiency in FVIII causes haemophilia A, a severe hereditary bleeding disorder. Intravenous administration of plasma-derived FVIII or recombinant FVIII concentrates restores normal coagulation in haemophilia A patients and is used as an effective therapy. In this work, we studied the biophysical properties of clinically potent recombinant FVIII forms: human FVIII full-length (FVIII-FL), human FVIII B-domain deleted (FVIII-BDD) and porcine FVIII-BDD bound to negatively charged phospholipid vesicles at near-physiological conditions. We used cryo-electron microscopy (Cryo-EM) as a direct method to evaluate the homogeneity and micro-organization of the protein-vesicle suspensions, which are important for FVIII therapeutic properties. Applying concurrent Cryo-EM, circular dichroism and dynamic light scattering studies to the three recombinant FVIII forms when bound to phospholipid vesicles revealed novel properties for their functional, membrane-bound state. The three FVIII constructs have similar activity, secondary structure distribution and bind specifically to negatively charged phospholipid membranes. Human and porcine FVIII-BDD induce strong aggregation of the vesicles, but the human FVIII-FL form does not. The proposed methodology is effective in characterizing and identifying differences in therapeutic recombinant FVIII membrane-bound forms near physiological conditions, because protein-containing aggregates are considered to be a factor in increasing the immunogenicity of protein therapeutics. This will provide better characterization and development of safer and more effective FVIII products with implications for haemophilia A treatment.


Asunto(s)
Factor VIII/química , Liposomas/química , Fusión de Membrana , Fosfatidilcolinas/química , Fosfolípidos/metabolismo , Microscopía por Crioelectrón/métodos , Factor VIII/inmunología , Humanos , Fosfolípidos/química , Proteínas Recombinantes , Dispersión de Radiación
3.
J Thromb Haemost ; 16(2): 303-315, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29197156

RESUMEN

Essentials Factor VIII inhibitors are the most serious complication in patients with hemophilia A. Aggregates in biopharmaceutical products are an immunogenic risk factor. Aggregates were identified in recombinant full-length factor VIII products. Aggregates in recombinant factor VIII products are identified by analytical ultracentrifugation. SUMMARY: Background The development of inhibitory anti-factor VIII antibodies is the most serious complication in the management of patients with hemophilia A. Studies have suggested that recombinant full-length FVIII is more immunogenic than plasma-derived FVIII, and that, among recombinant FVIII products, Kogenate is more immunogenic than Advate. Aggregates in biopharmaceutical products are considered a risk factor for the development of anti-drug antibodies. Objective To evaluate recombinant full-length FVIII products for the presence of aggregates. Methods Advate, Helixate and Kogenate were reconstituted to their therapeutic formulations, and subjected to sedimentation velocity (SV) analytical ultracentrifugation (AUC). Additionally, Advate and Kogenate were concentrated and subjected to buffer exchange by ultrafiltration to remove viscous cosolvents for the purpose of measuring s20,w values and molecular weights. Results The major component of all three products was a population of ~7.5 S heterodimers with a weight-average molecular weight of ~230 kDa. Helixate and Kogenate contained aggregates ranging from 12 S to at least 100 S, representing ≈ 20% of the protein mass. Aggregates greater than 12 S represented < 3% of the protein mass in Advate. An approximately 10.5 S aggregate, possibly representing a dimer of heterodimers, was identified in buffer-exchanged Advate and Kogenate. SV AUC analysis of a plasma-derived FVIII product was confounded by the presence of von Willebrand factor in molar excess over FVIII. Conclusions Aggregate formation has been identified in recombinant full-length FVIII products, and is more extensive in Helixate and Kogenate than in Advate. SV AUC is an important method for characterizing FVIII products.


Asunto(s)
Factor VIII/aislamiento & purificación , Agregado de Proteínas , Ultracentrifugación/métodos , Anticuerpos Neutralizantes , Cromatografía en Gel , Composición de Medicamentos , Factor VIII/inmunología , Humanos , Peso Molecular , Proteínas Recombinantes/aislamiento & purificación , Espectrofotometría Ultravioleta , Factor de von Willebrand/aislamiento & purificación
4.
J Thromb Haemost ; 16(9): 1779-1788, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29981270

RESUMEN

Essentials Inhibitor formation remains a challenging complication of hemophilia A care. The Bethesda assay is the primary method used for determining bleeding risk and management. Antibodies that block factor VIII binding to von Willebrand factor can increase FVIII clearance. Antibodies that increase clearance contribute to antibody pathogenicity. SUMMARY: Background The development of neutralizing anti-factor VIII (FVIII) antibodies remains a challenging complication of modern hemophilia A care. In vitro assays are the primary method used for quantifying inhibitor titers, predicting bleeding risk, and determining bleeding management. However, other mechanisms of inhibition are not accounted for in these assays, which may result in discrepancies between the inhibitor titer and clinical bleeding symptoms. Objectives To evaluate FVIII clearance in vivo as a potential mechanism for antibody pathogenicity and to determine whether increased FVIII dosing regimens correct the associated bleeding phenotype. Methods FVIII-/- or FVIII-/- /von Willebrand factor (VWF)-/- mice were infused with anti-FVIII mAbs directed against the FVIII C1, C2 or A2 domains, followed by infusion of FVIII. Blood loss via the tail snip bleeding model, FVIII activity and FVIII antigen levels were subsequently measured. Results Pathogenic anti-C1 mAbs that compete with VWF for FVIII binding increased the clearance of FVIII-mAb complexes in FVIII-/- mice but not in FVIII-/- /VWF-/- mice. Additionally, pathogenic anti-C2 mAbs that inhibit FVIII binding to VWF increased FVIII clearance in FVIII-/- mice. Anti-C1, anti-C2 and anti-A2 mAbs that do not inhibit VWF binding did not accelerate FVIII clearance. Infusion of increased doses of FVIII in the presence of anti-C1 mAbs partially corrected blood loss in FVIII-/- mice. Conclusions A subset of antibodies that inhibit VWF binding to FVIII increase the clearance of FVIII-mAb complexes, which contributes to antibody pathogenicity. This may explain differences in the bleeding phenotype observed despite factor replacement in some patients with hemophilia A and low-titer inhibitors.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Factor VIII/inmunología , Animales , Anticuerpos Heterófilos/administración & dosificación , Anticuerpos Heterófilos/inmunología , Anticuerpos Heterófilos/toxicidad , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/toxicidad , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/toxicidad , Epítopos/inmunología , Factor VIII/antagonistas & inhibidores , Factor VIII/farmacocinética , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Hemorragia/etiología , Concentración 50 Inhibidora , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Modelos Animales , Fenotipo , Dominios Proteicos , Enfermedades de von Willebrand , Factor de von Willebrand/metabolismo
5.
J Clin Invest ; 71(5): 1175-82, 1983 May.
Artículo en Inglés | MEDLINE | ID: mdl-6189858

RESUMEN

Acute iron loading of rats, by intraperitoneal administration of iron-dextran (500 mg Fe/kg body wt 18-20 h before killing) decreased by 30% the rate of conversion of 5-amino-[14C]levulinate ([14C]ALA) into heme as measured with a recently described procedure for liver homogenates (1981. Biochem. J. 198: 595-604). The decrease in conversion of labeled ALA into heme caused by iron loading was shown to be due to a 70-80% decrease in activity of ALA dehydrase. The decrease in activity of ALA dehydrase caused by iron loading was not associated with a decrease in hepatic concentrations of GSH, nor could it be reversed by addition of dithiothreitol, Zn2+ or chelators of Fe2+ and Fe3+. Addition of FeSO4, ferric citrate, or ferritin to homogenates of control liver had no effect of activity of ALA dehydrase. The decrease in activity of ALA dehydrase, caused by iron-dextran, was mirrored by a reciprocal increase in ALA synthase. Iron-dextran potentiated the induction of ALA synthase by allylisopropylacetamide. However, this potentiation could be dissociated from the decrease in ALA dehydrase caused by iron loading.


Asunto(s)
Dextranos/farmacología , Hemo/biosíntesis , Hierro/farmacología , Hígado/enzimología , Porfobilinógeno Sintasa/antagonistas & inhibidores , 5-Aminolevulinato Sintetasa/metabolismo , Alilisopropilacetamida/farmacología , Ácido Aminolevulínico/metabolismo , Animales , Sinergismo Farmacológico , Masculino , Protoporfirinas/biosíntesis , Ratas , Ratas Endogámicas , Uroporfirinógeno Descarboxilasa/metabolismo
6.
J Thromb Haemost ; 5(3): 512-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17181826

RESUMEN

BACKGROUND: Inhibitory antibodies (Abs) to factor VIII (FVIII inhibitors) constitute the most significant complication in the management of hemophilia A. The analysis of FVIII inhibitors is confounded by polyclonality and the size of FVIII. OBJECTIVES: The goal of this study was to dissect the polyclonal response to human FVIII in hemophilia A mice undergoing a dosage schedule that mimics human use. METHODS: Splenic B-cell hybridomas were obtained following serial i.v. injections of submicrogram doses of FVIII. Results of a novel, anti-FVIII domain-specific enzyme-linked immunosorbent assay were compared to Ab isotype and anti-FVIII inhibitory activity. RESULTS: The robust immune response resulted in the production of approximately 300 hybridomas per spleen. We characterized Abs from 506 hybridomas, representing the most comprehensive analysis of a protein antigen to date. Similar to the human response to FVIII, anti-A2 and anti-C2 Abs constituted the majority of inhibitors. A novel epitope was identified in the A2 domain by competition ELISA. Anti-A2 and anti-C2 Abs were significantly associated with IgG(1) and IgG(2a) isotypes, respectively. Because the IgG(2a) isotype is associated with enhanced Fc receptor-mediated effector mechanisms, this result suggests that anti-C2 Abs and inflammation may be linked. Additionally, we identified a novel class of Abs with dual specificity for the A1 and A3 domains. Forty per cent of the Abs had no detectable inhibitory activity, indicating that they are prominent and potentially pathologically significant. CONCLUSION: The expanded delineation of the humoral response to FVIII may lead to improved management of hemophilia A through mutagenesis of FVIII B-cell epitopes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos , Coagulantes/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Isotipos de Inmunoglobulinas/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/sangre , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Unión Competitiva , Línea Celular , Coagulantes/administración & dosificación , Cricetinae , Modelos Animales de Enfermedad , Esquema de Medicación , Ensayo de Inmunoadsorción Enzimática/métodos , Mapeo Epitopo/métodos , Factor VIII/administración & dosificación , Factor VIII/genética , Factor VIII/metabolismo , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Hibridomas/metabolismo , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/sangre , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/inmunología , Reproducibilidad de los Resultados , Bazo/citología , Bazo/metabolismo , Porcinos , Transfección
7.
J Thromb Haemost ; 15(9): 1867-1877, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28692141

RESUMEN

Essentials The mechanism for the auto-inhibition of von Willebrand factor (VWF) remains unclear. Hydrogen exchange of two VWF A1 fragments with disparate activities was measured and compared. Discontinuous residues flanking A1 form a structural module that blocks A1 binding to the platelet. Our results suggest a potentially unified model of VWF activation. Click to hear an ISTH Academy presentation on the domain architecture of VWF and activation by elongational flow by Dr Springer SUMMARY: Background How von Willebrand factor (VWF) senses and responds to shear flow remains unclear. In the absence of shear flow, VWF or its fragments can be induced to bind spontaneously to platelet GPIbα. Objectives To elucidate the auto-inhibition mechanism of VWF. Methods Hydrogen-deuterium exchange (HDX) of two recombinant VWF fragments expressed from baby hamster kidney cells were measured and compared. Results The shortA1 protein contains VWF residues 1261-1472 and binds GPIbα with a significantly higher affinity than the longA1 protein that contains VWF residues 1238-1472. Both proteins contain the VWF A1 domain (residues 1272-1458). Many residues in longA1, particularly those in the N- and C-terminal sequences flanking the A1 domain, and in helix α1, loops α1ß2 and ß3α2, demonstrated markedly reduced HDX compared with their counterparts in shortA1. The HDX-protected region in longA1 overlaps with the GPIbα-binding interface and is clustered with type 2B von Willebrand disease (VWD) mutations. Additional comparison with the HDX of denatured longA1 and ristocetin-bound longA1 indicates the N- and C-terminal sequences flanking the A1 domain form cooperatively an integrated autoinhibitory module (AIM) that interacts with the HDX-protected region. Binding of ristocetin to the C-terminal part of the AIM desorbs the AIM from A1 and enables longA1 binding to GPIbα. Conclusion The discontinuous AIM binds the A1 domain and prevents it from binding to GPIbα, which has significant implications for the pathogenesis of type 2B VWD and the shear-induced activation of VWF activity.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Factor de von Willebrand/metabolismo , Unión Competitiva , Medición de Intercambio de Deuterio , Humanos , Modelos Moleculares , Mutación , Fragmentos de Péptidos/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Factor de von Willebrand/química , Factor de von Willebrand/genética
8.
J Thromb Haemost ; 14(2): 346-55, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26588198

RESUMEN

UNLABELLED: ESSENTIALS: Anti-factor VIII (FVIII) inhibitory antibody formation is a severe complication in hemophilia A therapy. We genetically engineered and characterized a mouse model with complete deletion of the F8 coding region. F8(TKO) mice exhibit severe hemophilia, express no detectable F8 mRNA, and produce FVIII inhibitors. The defined background and lack of FVIII in F8(TKO) mice will aid in studying FVIII inhibitor formation. BACKGROUND: The most important complication in hemophilia A treatment is the development of inhibitory anti-Factor VIII (FVIII) antibodies in patients after FVIII therapy. Patients with severe hemophilia who express no endogenous FVIII (i.e. cross-reacting material, CRM) have the greatest incidence of inhibitor formation. However, current mouse models of severe hemophilia A produce low levels of truncated FVIII. The lack of a corresponding mouse model hampers the study of inhibitor formation in the complete absence of FVIII protein. OBJECTIVES: We aimed to generate and characterize a novel mouse model of severe hemophilia A (designated the F8(TKO) strain) lacking the complete coding sequence of F8 and any FVIII CRM. METHODS: Mice were created on a C57BL/6 background using Cre-Lox recombination and characterized using in vivo bleeding assays, measurement of FVIII activity by coagulation and chromogenic assays, and anti-FVIII antibody production using ELISA. RESULTS: All F8 exonic coding regions were deleted from the genome and no F8 mRNA was detected in F8(TKO) mice. The bleeding phenotype of F8(TKO) mice was comparable to E16 mice by measurements of factor activity and tail snip assay. Similar levels of anti-FVIII antibody titers after recombinant FVIII injections were observed between F8(TKO) and E16 mice. CONCLUSIONS: We describe a new C57BL/6 mouse model for severe hemophilia A patients lacking CRM. These mice can be directly bred to the many C57BL/6 strains of genetically engineered mice, which is valuable for studying the impact of a wide variety of genes on FVIII inhibitor formation on a defined genetic background.


Asunto(s)
Factor VIII/genética , Eliminación de Gen , Hemofilia A/genética , Hemostasis , Animales , Anticuerpos/sangre , Pruebas de Coagulación Sanguínea , Compuestos Cromogénicos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Factor VIII/inmunología , Factor VIII/metabolismo , Factor VIII/farmacología , Predisposición Genética a la Enfermedad , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Hemostasis/efectos de los fármacos , Hemostasis/genética , Hemostáticos/inmunología , Hemostáticos/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Índice de Severidad de la Enfermedad
9.
Biochim Biophys Acta ; 1080(3): 245-51, 1991 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-1954232

RESUMEN

Relationships between activities of delta-aminolevulinate synthase and heme oxygenase, respectively the rate-limiting enzymes of heme biosynthesis and degradation, have been studied in chick embryo liver cell cultures following exposure of the cultures to glutethimide and iron, a combination known to produce a synergistic induction of both enzymes. In time-course experiments, synergistic induction of heme oxygenase activity by glutethimide and iron preceded that of delta-aminolevulinate synthase by 4 h. Effects of selective inhibitors of both heme synthesis and degradation have also been studied with respect to effects on delta-aminolevulinate synthase and heme oxygenase activities. The synergistic induction of heme oxygenase by glutethimide and iron appears to be dependent upon cellular heme synthesis because addition of inhibitors of heme biosynthesis, 4,6-dioxoheptanoic acid or N-methyl-mesoporphyrin abolishes this synergistic induction. Exposure of cultures to tin-mesoporphyrin, a potent inhibitor of heme oxygenase, prevented the synergistic induction of delta-aminolevulinate synthase produced by glutethimide and iron, or, when added after induction was already established, promptly halted any further induction. These results suggest that the level of activity of heme oxygenase can reciprocally modulate intracellular heme levels and thus activity of delta-aminolevulinate synthase.


Asunto(s)
5-Aminolevulinato Sintetasa/efectos de los fármacos , Glutetimida/farmacología , Hemo Oxigenasa (Desciclizante)/efectos de los fármacos , Hierro/farmacología , 5-Aminolevulinato Sintetasa/biosíntesis , Animales , Células Cultivadas , Embrión de Pollo , Sinergismo Farmacológico , Inducción Enzimática/efectos de los fármacos , Compuestos Férricos/farmacología , Hemo/farmacología , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hígado/embriología , Hígado/enzimología , Metaloporfirinas/farmacología , Ácido Nitrilotriacético/análogos & derivados , Ácido Nitrilotriacético/farmacología
10.
Invest Radiol ; 11(3): 182-6, 1976.
Artículo en Inglés | MEDLINE | ID: mdl-947222

RESUMEN

The water soluble radiopaque medium, metrizamide (Amipaque) was introduced into the lumbar subarachnoid space in chloralose anesthetized cats at a standard volume of 0.35 cc/kg in concentrations of 300 mgI/cc to 500 mgI/cc during EMG recording. These animals did not differ from controls which received cerebrospinal fluid under otherwise identical conditions; both groups usually showed some mild and occasional muscle fasciculations or mild spasms. Treatment with metrizamide appeared to be a less deleterious procedure than that using hyperosmotic sucrose (1.32 M) as judged from EMG records. In contrast, equivalent amounts of neglumine iothalamate produced frank convulsions in 7 of 15 cases and a range of hyperirritability in the remaining 8.


Asunto(s)
Medios de Contraste/toxicidad , Yodobenzoatos/toxicidad , Metrizamida/toxicidad , Convulsiones/inducido químicamente , Anestesia , Animales , Presión Sanguínea/efectos de los fármacos , Gatos , Cloralosa , Medios de Contraste/administración & dosificación , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Inyecciones Espinales , Yotalamato de Meglumina/toxicidad , Masculino , Metrizamida/administración & dosificación , Respiración/efectos de los fármacos , Espacio Subaracnoideo
11.
Arch Pathol Lab Med ; 103(1): 21-9, 1979 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-215100

RESUMEN

To understand better the intracellular iron distribution and metabolic consequences of chronic hepatic iron overload, rats were given large doses of iron dextran or ferric citrate intraperitoneally. They accumulated large quantities of iron within Kupffer cells and hepatocytes. The relative subcellular iron distributions were similar in controls and iron-loaded rats, despite a ten- to 20-fold difference in hepatic iron concentration. Electron microscopy of whole liver and subcellular particulate fractions suggested that iron was present in highest concentration in lysosomes, which were rendered more labile by its presence. Nevertheless, quantitative iron determinations on all subcellular fractions, obtained by two preparative methods, showed that most of the iron was present in the "soluble" fraction. The amount of iron in the "microsomal" fraction varied, depending on the techniques used for preparation of this fraction. Cytochrome P-450 and total heme concentrations were decreased 40% to 50% in microsomes isolated from iron-loaded livers.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Compuestos Férricos/administración & dosificación , Complejo Hierro-Dextran/administración & dosificación , Hierro/administración & dosificación , Hierro/metabolismo , Hígado/citología , Microsomas Hepáticos/enzimología , Animales , Citocromos/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Glucuronidasa/metabolismo , Hemo/metabolismo , Macrófagos del Hígado/citología , Hígado/metabolismo , Masculino , Mitocondrias Hepáticas/enzimología , Ratas
12.
J Thromb Haemost ; 11(12): 2128-36, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24152306

RESUMEN

BACKGROUND: The development of anti-factor VIII antibodies (inhibitors) is a significant complication in the management of patients with hemophilia A, leading to significant increases in morbidity and treatment cost. Using a panel of mAbs against different epitopes on FVIII, we have recently shown that epitope specificity, inhibitor kinetics and time to maximum inhibition are more important than inhibitor titer in predicting responses to FVIII and the combination of FVIII and recombinant FVIIa. In particular, a subset of high-titer inhibitors responded to high-dose FVIII, which would not be predicted on the basis of their inhibitor titer alone. Thus, the ability to quickly map the epitope spectrum of patient plasma with a clinically feasible assay may fundamentally change how clinicians approach the treatment of high-titer inhibitor patients. OBJECTIVES: To map the epitopes of anti-FVIII mAbs, three of which are classic inhibitors and one of which is a non-classic inhibitor, by the use of hydrogen-deuterium exchange coupled with mass spectrometry (HDX-MS). METHODS: The binding epitopes of four mAbs targeting the FVIII C2 domain were mapped with HDX-MS. RESULTS: The epitopes determined with HDX-MS are consistent with those obtained earlier through structural characterization and antibody competition assays. In addition, classic and non-classic inhibitor epitopes could be distinguished by the use of a limited subset of C2 domain-derived peptic fragments. CONCLUSION: Our results demonstrate the effectiveness and robustness of the HDX-MS method for epitope mapping, and suggest a potential role of rapid mapping of FVIII inhibitor epitopes in facilitating individualized treatment of inhibitor patients.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Mapeo Epitopo , Factor VIII/inmunología , Espectrometría de Masas/métodos , Cromatografía Líquida de Alta Presión , Deuterio , Humanos , Hidrógeno , Proteínas Recombinantes/inmunología
15.
J Thromb Haemost ; 7(4): 658-64, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19187078

RESUMEN

OBJECTIVE: The pathogenicity of anti-human factor (F) VIII monoclonal antibodies (MAbs) was tested in a murine bleeding model. METHODS: MAbs were injected into the tail veins of hemophilia A mice to a peak plasma concentration of 60 nm, followed by injection of human B domain-deleted FVIII at 180 U kg(-1), producing peak plasma concentrations of approximately 2 nm. At 2 h, blood loss following a 4-mm tail snip was measured. The following MAbs were tested: (i) 4A4, a type I anti-A2 FVIII inhibitor, (ii) I54 and 1B5, classical type I anti-C2 inhibitors, (iii) 2-77 and B45, non-classical type II anti-C2 inhibitors, and (iv) 2-117, a non-classical anti-C2 MAb with inhibitory activity less than 0.4 Bethesda Units per mg IgG. RESULTS: All MAbs except 2-117 produced similar amounts of blood loss that were significantly greater than control mice injected with FVIII alone. Increasing the dose of FVIII to 360 U kg(-1) overcame the bleeding diathesis produced by the type II MAbs 2-77 and B45, but not the type I antibodies, 4A4, I54, and 1B5. These results were consistent with the in vitro Bethesda assay in which 4A4 completely inhibited both 1 U mL(-1) and 3 U mL(-1) FVIII, while there was 40% residual activity at saturating concentrations of 2-77 at either concentration of FVIII. CONCLUSIONS: For patients with an inhibitor response dominated by non-classical anti-C2 antibodies both the in vivo and in vitro results suggest that treatment with high-dose FVIII rather than bypassing agents may be warranted.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Epítopos/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Hemorragia/inmunología , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/sangre , Modelos Animales de Enfermedad , Factor VIII/administración & dosificación , Humanos , Ratones
16.
Eur J Biochem ; 189(1): 155-66, 1990 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-2158889

RESUMEN

A major inducible form of heme oxygenase (EC 1.14.99.3) was purified from liver microsomes of chicks pretreated with cadmium chloride. The purification involved solubilization of microsomes with Emulgen 913 and sodium cholate, followed by DEAE-Sephacel, carboxymethyl-cellulose (CM-52) and hydroxyapatite chromatography, and FPLC through Superose 6 and 12 columns operating in series. The final product gave a single band on silver-stained SDS/polyacrylamide gels (Mr = 33,000). Optimal conditions for measurement of activity of solubilized heme oxygenase were studied. In a reconstituted system containing purified heme oxygenase, NADPH-cytochrome reductase, biliverdin reductase and NADPH, the Km for free heme was 3.8 +/- 0.5 microM; for heme in the presence of bovine serum albumin (5 mol heme/3 mol albumin) the Km was 5.0 +/- 0.8 microM; and the Km for NADPH was 6.1 +/- 0.4 microM (all values mean +/- SD, n = 3). Oxygen concentration as low as 15 microM, with saturating concentrations of heme and NADPH, did not affect the reaction rate, indicating that the supply of oxygen is not involved in the physiological regulation of activity of the enzyme. The pH optimum of the reaction was 7.4; at 37 degrees C, the apparent Vmax was 580 +/- 44 nmol biliverdin.(mg protein)-1.min-1 and the molecular activity was 19.2 min-1. Biliverdin IXa was the sole biliverdin isomer formed. In the presence of purified biliverdin reductase, biliverdin was converted quantitatively to bilirubin. Addition of catalase to the reconstituted system decreased the breakdown of heme to non-biliverdin products and led to nearly stoichiometric conversion of heme to biliverdin. Activity of the enzyme in the reconstituted system was inhibited by metalloporphyrins in the following order of decreasing potency: tin mesoporphyrin greater than tin protoporphyrin greater than zinc protoporphyrin greater than manganese protoporphyrin greater than cobalt protoporphyrin. Protoporphyrin (3.3 or 6.6 microM) (and several other porphyrins) and metallic ions (100 microM) alone had little if any inhibitory effect, except for Hg2+ which inhibited by 67% at 10 microM and totally at 15 microM. Following partial cleavage, fragments of the purified enzyme were sequenced. Comparison of sequences to those derived from cDNA sequences for the major inducible rat and human heme oxygenase showed 69% and 76% similarities, respectively. The histidine residue at position 132 of rat heme oxygenase-1 and the residues (Lys128-Arg136) flanking His132 were conserved in all three enzymes, as well as in the corresponding portion of a fourth less highly similar rat enzyme, heme oxygenase-2.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Hemo Oxigenasa (Desciclizante)/aislamiento & purificación , Microsomas Hepáticos/enzimología , Oxigenasas de Función Mixta/aislamiento & purificación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Secuencia de Aminoácidos , Animales , Cationes , Bovinos , Pollos , Cromatografía , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Durapatita , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Hidroxiapatitas , Cinética , Masculino , Datos de Secuencia Molecular , Peso Molecular , NADPH-Ferrihemoproteína Reductasa/aislamiento & purificación , NADPH-Ferrihemoproteína Reductasa/metabolismo , Oxidorreductasas/aislamiento & purificación , Oxidorreductasas/metabolismo , Porfirinas/farmacología , Ratas , Homología de Secuencia de Ácido Nucleico
17.
Biochem J ; 250(1): 189-96, 1988 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-3355510

RESUMEN

We studied drug- and metal-mediated increases in activity of haem oxygenase, the rate-controlling enzyme for haem breakdown, in chick-embryo hepatocytes in ovo and in primary culture. Phenobarbitone and phenobarbitone-like drugs (glutethimide, mephenytoin), which are known to increase concentrations of an isoform of cytochrome P-450 in chick-embryo hepatocytes, were found to increase activities of haem oxygenase as well. In contrast, 20-methylcholanthrene, which increases the concentration of a different isoform of cytochrome P-450, had no effect on activity of haem oxygenase. Inhibitors of haem synthesis, 4,6-dioxoheptanoic acid or desferrioxamine, prevented drug-mediated induction of both cytochrome P-450 and haem oxygenase in embryo hepatocytes in ovo or in culture. Addition of haem restored induction of both enzymes. These results are interpreted to indicate that phenobarbitone and its congeners induce haem oxygenase by increasing hepatic haem formation. In contrast, increases in haem oxygenase activity by metals such as cobalt, cadmium and iron were not dependent on increased haem synthesis and were not inhibited by 4,6-dioxoheptanoic acid. We conclude that (1) induction of hepatic haem oxygenase activity by phenobarbitone-type drugs is due to increased haem formation, and (2) induction of haem oxygenase by drugs and metals occurs by different mechanisms.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo/metabolismo , Hígado/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Animales , Cationes Bivalentes/farmacología , Células Cultivadas , Embrión de Pollo , Cicloheximida/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Dactinomicina/farmacología , Inducción Enzimática/efectos de los fármacos , Glutetimida/farmacología , Hígado/efectos de los fármacos , Mefenitoína/farmacología , Metilcolantreno/farmacología , Fenobarbital/farmacología , Porfirinas/metabolismo
18.
Biochem J ; 279 ( Pt 3): 849-54, 1991 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-1953681

RESUMEN

Monospecific polyclonal rabbit antibodies to a purified form of haem oxygenase of chick liver, showing sequence similarity to mammalian haem oxygenase-1, were raised and used to study characteristics of the oxygenase. The antibodies inhibited activity of the purified oxygenase, but not other enzyme components (NADPH:cytochrome reductase and biliverdin reductase) of the standard assay mixture of haem oxygenase. In addition, the antibodies inhibited activity of haem oxygenase in microsomes (microsomal fractions) from Cd(2+)-treated chick liver, spleen, testis and brain. Western (immuno-) blots of microsomal proteins of selected organs from chick, rat and man, and homogenates of chick-embryo liver-cell cultures, probed with the antibodies, showed a major protein with a molecular mass of 33-34 kDa and a lower-molecular-mass protein (28-29 kDa) of variable intensity. Studies with trypsin and selected proteinase inhibitors established that the smaller peptide was a proteolytic product of the larger. Treatment of chick-embryo liver-cell cultures with CdCl2, a potent inducer of haem oxygenase, increased the degree of proteinase-mediated cleavage of the 33 kDa protein to the lower-molecular-mass form. These results indicate that, under at least some conditions, such cultures should be homogenized in the presence of trypsin inhibitor to prevent proteolytic degradation of the enzyme and allow maximal expression of haem oxygenase activity. The antibodies also reacted with haem oxygenase from spleen, testis and brain of both chicks and rats, and the spleen of humans. A method for quantifying the amount of haem oxygenase protein was developed with use of slot-blots and laser densitometry; linearity was observed from 0 to 5 ng of haem oxygenase protein per slot, and the method was applied to sonicated cultured chick-embryo liver cells treated with Cd2+ (0.3 mM) or iron plus glutethimide. In both cases, increases in enzyme activity were of similar magnitude to increases in amounts of enzyme protein. Approximate amounts of haem oxygenase protein in microsomes of several organs from intact animals could also be estimated by the use of slot-blot-laser densitometry, and the amounts measured were increased by the addition of purified haem oxygenase to the microsomal preparations. Results of these studies indicated that haem oxygenase-1 could be detected in microsomes from all chick or rat organs studied, including testis and brain.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Anticuerpos/química , Hemo Oxigenasa (Desciclizante)/química , Microsomas Hepáticos/enzimología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Animales , Cadmio/farmacología , Embrión de Pollo , Pollos , Inducción Enzimática , Glutetimida/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/biosíntesis , Humanos , Immunoblotting , Inmunoglobulina G/fisiología , Hierro/farmacología , Masculino , NADH Deshidrogenasa/química , NADH Deshidrogenasa/inmunología , Oxidorreductasas/química , Oxidorreductasas/inmunología , Inhibidores de Proteasas/farmacología , Conejos , Ratas
19.
J Biol Chem ; 269(1): 593-8, 1994 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-8276857

RESUMEN

The activation of factor VIII (fVIII) by thrombin is associated with heavy chain cleavages at Arg372 and Arg740 and light chain cleavage at Arg1689. In a defined, plasma-free assay of fVIII activation and at physiological ionic strength and pH, heparin inhibited the rate of activation of either human or porcine fVIII by thrombin in either the presence or absence of von Willebrand factor (vWf). The inhibitory effect of heparin was associated with inhibition of all three thrombin-catalyzed bond cleavages. At plasma concentrations of fVIII (approximately 1 nM) and vWf (approximately 35 nM), the rate of fVIII activation was inhibited by 50% at approximately 0.1 unit/ml heparin, which is below the normal range of heparin concentrations in plasma during therapeutic anticoagulation (0.2-0.7 unit/ml). We propose that, in addition to catalyzing the inhibition of thrombin and other intrinsic pathway coagulation proteases by antithrombin, heparin functions as an anticoagulant by direct inhibition of the activation of the fVIII-vWf complex by thrombin.


Asunto(s)
Factor VIII/metabolismo , Heparina/farmacología , Trombina/farmacología , Factor de von Willebrand/metabolismo , Animales , Catálisis , Fibrinógeno/farmacología , Heparina/metabolismo , Humanos , Hidrólisis , Sefarosa/metabolismo , Porcinos , Trombina/antagonistas & inhibidores
20.
Blood ; 88(11): 4209-14, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8943856

RESUMEN

The cDNA corresponding to 137 bp of the 5' untranslated region, the signal peptide, and the A1, A3, C1, and C2 domains of porcine factor VIII (fVIII) have been cloned and sequenced. Along with previously determined sequences of the porcine fVIII B domain and the A2 domain, this completes the sequence determination of the cDNA corresponding to the translated protein. Alignments of the derived amino acid sequence of porcine fVIII with human and murine fVIII indicate that the A1, A2, A3, C1, and C2 domains are more conserved than the B domains or the proteolytic cleavage peptides corresponding to residues 337-372 and 1649-1689. The knowledge of the porcine fVIII cDNA may be useful to understand functional and immunological differences between human and porcine fVIII and may lead to improved fVIII replacement products for hemophilia. A patients through the development of recombinant porcine fVIII or hybrid human/porcine fVIII derivatives.


Asunto(s)
ADN Complementario/genética , Factor VIII/genética , Porcinos/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Epítopos/química , Epítopos/inmunología , Factor VIII/inmunología , Genes , Humanos , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Señales de Clasificación de Proteína/química , Alineación de Secuencia , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA