Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Hematology Am Soc Hematol Educ Program ; 2018(1): 371-376, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30504334

RESUMEN

Atypical hemolytic uremic syndrome (aHUS); hemolysis, elevated liver function tests, and low platelets syndrome; and transplant-associated thrombotic microangiopathy are related conditions, in that many patients harbor germline heterozygous mutations in genes that regulate the alternative pathway of complement (APC). Penetrance is variable because development of clinically significant disease appears to require supervention of a process such as inflammation. Complement activation on the endothelial surfaces leads to endothelial damage, platelet consumption, microthrombi, and a mechanical hemolytic anemia with schistocytes. Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal hematopoietic disease caused by expansion of a stem cell that harbors a somatic mutation in PIGA PIGA mutant blood cells are deficient in the complement regulator proteins CD55 and CD59, making them susceptible to intravascular hemolysis due to a failure to regulate the APC on erythrocytes. Eculizumab is a monoclonal antibody that binds to C5 and inhibits terminal complement by interfering with the cleavage of C5 by the C5 convertases. The drug is approved by the US Food and Drug Administration for the treatment of aHUS and PNH; however, a new generation of complement inhibitors that block C5 and other components of the complement cascade is showing promise in preclinical and clinical trials.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico , Hemoglobinuria Paroxística , Mutación , Síndrome Hemolítico Urémico Atípico/sangre , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/genética , Antígenos CD55/sangre , Antígenos CD55/genética , Antígenos CD59/sangre , Antígenos CD59/genética , Complemento C5/antagonistas & inhibidores , Complemento C5/genética , Complemento C5/metabolismo , C5 Convertasa de la Vía Alternativa del Complemento/antagonistas & inhibidores , C5 Convertasa de la Vía Alternativa del Complemento/genética , C5 Convertasa de la Vía Alternativa del Complemento/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Vía Alternativa del Complemento/genética , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/tratamiento farmacológico , Hemoglobinuria Paroxística/genética , Hemólisis/efectos de los fármacos , Hemólisis/genética , Humanos , Proteínas de la Membrana/sangre , Proteínas de la Membrana/genética , Penetrancia
2.
BMC Biol ; 13: 93, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26552476

RESUMEN

BACKGROUND: Complement is a large protein network in plasma that is crucial for human immune defenses and a major cause of aberrant inflammatory reactions. The C5 convertase is a multi-molecular protease complex that catalyses the cleavage of native C5 into its biologically important products. So far, it has been difficult to study the exact molecular arrangement of C5 convertases, because their non-catalytic subunits (C3b) are covalently linked to biological surfaces through a reactive thioester. Through development of a highly purified model system for C5 convertases, we here aim to provide insights into the surface-specific nature of these important protease complexes. RESULTS: Alternative pathway (AP) C5 convertases were generated on small streptavidin beads that were coated with purified C3b molecules. Site-specific biotinylation of C3b via the thioester allowed binding of C3b in the natural orientation on the surface. In the presence of factor B and factor D, these C3b beads could effectively convert C5. Conversion rates of surface-bound C3b were more than 100-fold higher than fluid-phase C3b, confirming the requirement of a surface. We determine that high surface densities of C3b, and its attachment via the thioester, are essential for C5 convertase formation. Combining our results with molecular modeling explains how high C3b densities may facilitate intermolecular interactions that only occur on target surfaces. Finally, we define two interfaces on C5 important for its recognition by surface-bound C5 convertases. CONCLUSIONS: We establish a highly purified model that mimics the natural arrangement of C5 convertases on a surface. The developed model and molecular insights are essential to understand the molecular basis of deregulated complement activity in human disease and will facilitate future design of therapeutic interventions against these critical enzymes in inflammation.


Asunto(s)
Complemento C3b/metabolismo , C5 Convertasa de la Vía Alternativa del Complemento/química , Catálisis , C5 Convertasa de la Vía Alternativa del Complemento/metabolismo , Humanos , Cinética , Microesferas , Modelos Químicos , Estreptavidina/química
3.
J Am Soc Nephrol ; 25(9): 2053-65, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24652797

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a genetic ultrarare renal disease associated with overactivation of the alternative pathway of complement. Four gain-of-function mutations that form a hyperactive or deregulated C3 convertase have been identified in Factor B (FB) ligand binding sites. Here, we studied the functional consequences of 10 FB genetic changes recently identified from different aHUS cohorts. Using several tests for alternative C3 and C5 convertase formation and regulation, we identified two gain-of-function and potentially disease-relevant mutations that formed either an overactive convertase (M433I) or a convertase resistant to decay by FH (K298Q). One mutation (R178Q) produced a partially cleaved protein with no ligand binding or functional activity. Seven genetic changes led to near-normal or only slightly reduced ligand binding and functional activity compared with the most common polymorphism at position 7, R7. Notably, none of the algorithms used to predict the disease relevance of FB mutations agreed completely with the experimental data, suggesting that in silico approaches should be undertaken with caution. These data, combined with previously published results, suggest that 9 of 15 FB genetic changes identified in patients with aHUS are unrelated to disease pathogenesis. This study highlights that functional assessment of identified nucleotide changes in FB is mandatory to confirm disease association.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Factor B del Complemento/genética , Mutación , Sustitución de Aminoácidos , Sitios de Unión/genética , C3 Convertasa de la Vía Alternativa del Complemento/química , C3 Convertasa de la Vía Alternativa del Complemento/genética , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Complemento C3b/metabolismo , C5 Convertasa de la Vía Alternativa del Complemento/química , C5 Convertasa de la Vía Alternativa del Complemento/genética , C5 Convertasa de la Vía Alternativa del Complemento/metabolismo , Factor B del Complemento/química , Factor B del Complemento/metabolismo , Vía Alternativa del Complemento/genética , Simulación por Computador , Frecuencia de los Genes , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Modelos Moleculares , Complejos Multiproteicos/química , Polimorfismo Genético , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
J Biol Chem ; 284(16): 10473-9, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19196712

RESUMEN

Amplification of the complement cascade through the alternative pathway can lead to excessive inflammation. Targeting C3b, a component central to the alternative pathway of complement, provides a powerful approach to inhibit complement-mediated immune responses and tissue injury. In the present study, phage display technology was employed to generate an antibody that selectively recognizes C3b but not the non-activated molecule C3. The crystal structure of C3b in complex with a Fab fragment of this antibody (S77) illustrates the structural basis for this selectivity. Cleavage of C3 to C3b results in a plethora of structural changes within C3, including the rearrangement of macroglobulin domain 6 enabling binding of S77 to the adjacent macroglobulin domain 7 domain. S77 blocks binding of factor B to C3b inhibiting the first step in the formation of the alternative pathway C3 convertase. In addition, S77 inhibits C5 binding to C3b. This results in significantly reduced formations of anaphylatoxins and membrane-attack complexes. This study for the first time demonstrates the structural basis for complement inhibition by a C3b-selective antibody and provides insights into the molecular mechanisms of alternative pathway complement activation.


Asunto(s)
Anticuerpos , Complemento C3b/inmunología , Vía Alternativa del Complemento/fisiología , Fragmentos Fab de Inmunoglobulinas , Conformación Proteica , Animales , Anticuerpos/química , Anticuerpos/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Complemento C3b/química , Complemento C3b/genética , Complemento C3b/metabolismo , C5 Convertasa de la Vía Alternativa del Complemento/metabolismo , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Estabilidad de Enzimas , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Macaca mulatta , Modelos Moleculares , Datos de Secuencia Molecular , Biblioteca de Péptidos , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Receptores de Complemento 3b/química , Receptores de Complemento 3b/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...