Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-36853837

RESUMEN

Complement is a major innate defense system that protects the intravascular space from microbial invasion. Complement activation results in the assembly of C3 convertases, serine proteases that cleave complement protein C3, generating bioactive fragments C3a and C3b. The complement response is rapid and robust, largely due to a positive feedback regulatory loop mediated by alternative pathway (AP) C3 convertase. C3 nephritic factors (C3NEFs) are autoantibodies that stabilize AP convertase, resulting in uncontrolled C3 cleavage, which, in principle, can promote critical tissue injury similar to that seen in certain renal conditions. Investigations of C3NEFs are hampered by a challenging issue: each C3NEF is derived from a different donor source, and there is no method to compare one C3NEF to another. We have identified a widely available mouse anti-C3 mAb that, similar to many C3NEFs, can stabilize functional AP convertase in a form resistant to decay acceleration by multiple complement regulators. The antibody requires the presence of properdin to confer convertase stability, and hampers the activity of Salp20, a tic salivary protein that accelerates convertase dissociation by displacing properdin from the convertase complex. This mAb can serve as an urgently needed standard for the investigation of C3NEFs. This study also provides novel insights into the dynamics of AP convertase.


Asunto(s)
Anticuerpos Monoclonales , Factor Nefrítico del Complemento 3 , Animales , Ratones , Properdina , Autoanticuerpos
2.
Mol Immunol ; 69: 62-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26675068

RESUMEN

Complement, a major component of innate immunity, presents a rapid and robust defense of the intravascular space. While regulatory proteins protect host cells from complement attack, when these measures fail, unrestrained complement activation may trigger self-tissue injury, leading to pathologic conditions. Of the three complement activation pathways, the alternative pathway (AP) in particular has been implicated in numerous disease and injury states. Consequently, the AP components represent attractive targets for therapeutic intervention. The common hard-bodied ticks from the family Ixodidae derive nourishment from the blood of their mammalian hosts. During its blood meal the tick is exposed to host immune effectors, including the complement system. In defense, the tick produces salivary proteins that can inhibit host immune functions. The Salp20 salivary protein of Ixodes scapularis inhibits the host AP pathway by binding properdin and dissociating C3bBbP, the active C3 convertase. In these studies we examined Salp20 activity in various complement-mediated pathologies. Our results indicate that Salp20 can inhibit AP-dependent pathogenesis in the mouse. Its efficacy may be part in due to synergic effects it provides with the endogenous AP regulator, factor H. While Salp20 itself would be expected to be highly immunogenic and therefore inappropriate for therapeutic use, its emergence speaks for the potential development of a non-immunogenic Salp20 mimic that replicates its anti-properdin activity.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Ixodes/inmunología , Infestaciones por Garrapatas/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas y Péptidos Salivales/inmunología , Transfección
3.
Nanomedicine ; 11(3): 601-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25652900

RESUMEN

High-relaxivity T1-weighted (T1w) MR molecular imaging nanoparticles typically present high surface gadolinium payloads that can elicit significant acute complement activation (CA). The objective of this research was to develop a high T1w contrast nanoparticle with improved safety. We report the development, optimization, and characterization of a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC; 138±10 (Dav)/nm; PDI: 0.06; zeta: -27±2 mV). High r1 particulate relaxivity with minute additions of Gd-DOTA-lipid conjugate to the MnOL nanocolloid surface achieved an unexpected paramagnetic synergism. This hybrid MnOL-Gd NC provided optimal MR TSE signal intensity at 5 nM/voxel and lower levels consistent with the level expression anticipated for sparse biomarkers, such as neovascular integrins. MnOL NC produced optimal MR TSE signal intensity at 10 nM/voxel concentrations and above. Importantly, MnOL-Gd NC avoided acute CA in vitro and in vivo while retaining minimal transmetallation risk. From the clinical editor: The authors developed a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) in this study. These were used as a high-relaxivity paramagnetic MR molecular imaging agent in experimental models. It was shown that MnOL-Gd NC could provide high T1w MR contrast for targeted imaging. As the level of gadolinium used was reduced, there was also reduced risk of systemic side effects from complement activation.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Medios de Contraste , Gadolinio , Imagen por Resonancia Magnética , Manganeso , Nanopartículas , Animales , Biomarcadores/sangre , Coloides , Medios de Contraste/efectos adversos , Medios de Contraste/química , Medios de Contraste/farmacología , Evaluación Preclínica de Medicamentos , Gadolinio/efectos adversos , Gadolinio/química , Gadolinio/farmacología , Manganeso/efectos adversos , Manganeso/química , Manganeso/farmacología , Ratones , Nanopartículas/efectos adversos , Nanopartículas/química
4.
Artículo en Inglés | MEDLINE | ID: mdl-25723276

RESUMEN

The complement alternative pathway (AP) is a major contributor to a broad and growing spectrum of diseases that includes age-related macular degeneration, atypical hemolytic uremic syndrome, and preeclampsia. As a result, there is much interest in the therapeutic disruption of AP activity. Properdin, the only positive regulator of the AP, is a particularly promising AP target. Several issues need to be clarified before the potential for properdin-directed therapy can be realized. In this report we use a portion of the mouse properdin protein, expressed in a bacterial system, to raise rabbit polyclonal and hamster monoclonal antibodies that block properdin-dependent pathogenesis. These antibodies, when employed with AP-dependent mouse disease models, can help evaluate the feasibility of properdin-directed therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Aneurisma de la Aorta Abdominal/prevención & control , Vía Alternativa del Complemento/efectos de los fármacos , Inmunosupresores/farmacología , Properdina/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/biosíntesis , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Cricetinae , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Expresión Génica , Inmunosupresores/metabolismo , Ratones , Ratones Endogámicos C57BL , Elastasa Pancreática , Properdina/genética , Properdina/inmunología , Conejos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología
5.
Nanomedicine ; 10(3): 651-60, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24211337

RESUMEN

Nanoparticles offer new options for medical diagnosis and therapeutics with their capacity to specifically target cells and tissues with imaging agents and/or drug payloads. The unique physical aspects of nanoparticles present new challenges for this promising technology. Studies indicate that nanoparticles often elicit moderate to severe complement activation. Using human in vitro assays that corroborated the mouse in vivo results we previously presented mechanistic studies that define the pathway and key components involved in modulating complement interactions with several gadolinium-functionalized perfluorocarbon nanoparticles (PFOB). Here we employ a modified in vitro hemolysis-based assay developed in conjunction with the mouse in vivo model to broaden our analysis to include PFOBs of varying size, charge and surface chemistry and examine the variations in nanoparticle-mediated complement activity between individuals. This approach may provide the tools for an in-depth structure-activity relationship study that will guide the eventual development of biocompatible nanoparticles. FROM THE CLINICAL EDITOR: Unique physical aspects of nanoparticles may lead to moderate to severe complement activation in vivo, which represents a challenge to clinical applicability. In order to guide the eventual development of biocompatible nanoparticles, this team of authors report a modified in vitro hemolysis-based assay developed in conjunction with their previously presented mouse model to enable in-depth structure-activity relationship studies.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Fluorocarburos/inmunología , Hemólisis/efectos de los fármacos , Nanopartículas/metabolismo , Animales , Fluorocarburos/química , Humanos , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Tamaño de la Partícula
6.
J Clin Invest ; 123(5): 2218-30, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23619360

RESUMEN

During complement activation the C3 protein is cleaved, and C3 activation fragments are covalently fixed to tissues. Tissue-bound C3 fragments are a durable biomarker of tissue inflammation, and these fragments have been exploited as addressable binding ligands for targeted therapeutics and diagnostic agents. We have generated cross-reactive murine monoclonal antibodies against human and mouse C3d, the final C3 degradation fragment generated during complement activation. We developed 3 monoclonal antibodies (3d8b, 3d9a, and 3d29) that preferentially bind to the iC3b, C3dg, and C3d fragments in solution, but do not bind to intact C3 or C3b. The same 3 clones also bind to tissue-bound C3 activation fragments when injected systemically. Using mouse models of renal and ocular disease, we confirmed that, following systemic injection, the antibodies accumulated at sites of C3 fragment deposition within the glomerulus, the renal tubulointerstitium, and the posterior pole of the eye. To detect antibodies bound within the eye, we used optical imaging and observed accumulation of the antibodies within retinal lesions in a model of choroidal neovascularization (CNV). Our results demonstrate that imaging methods that use these antibodies may provide a sensitive means of detecting and monitoring complement activation-associated tissue inflammation.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/inmunología , Activación de Complemento , Complemento C3d/inmunología , Animales , Biomarcadores/metabolismo , Neovascularización Coroidal/metabolismo , Convertasas de Complemento C3-C5/inmunología , Complemento C3d/fisiología , Epítopos/inmunología , Humanos , Inflamación , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Proteínas Recombinantes/inmunología , Bazo/citología , Resonancia por Plasmón de Superficie
7.
J Biol Chem ; 286(41): 35725-35732, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21862585

RESUMEN

Factor B is a zymogen that carries the catalytic site of the complement alternative pathway C3 convertase. During convertase assembly, factor B associates with C3b and Mg(2+) forming a pro-convertase C3bB(Mg(2+)) that is cleaved at a single factor B site by factor D. In free factor B, a pair of salt bridges binds the Arg(234) side chain to Glu(446) and to Glu(207), forming a double latch structure that sequesters the scissile bond (between Arg(234) and Lys(235)) and minimizes its unproductive cleavage. It is unknown how the double latch is released in the pro-convertase. Here, we introduce single amino acid substitutions into factor B that preclude one or both of the Arg(234) salt bridges, and we examine their impact on several different pro-convertase complexes. Our results indicate that loss of the Arg(234)-Glu(446) salt bridge partially stabilizes C3bB(Mg(2+)). Loss of the Arg(234)-Glu(207) salt bridge has lesser effects. We propose that when factor B first associates with C3b, it bears two intact Arg(234) salt bridges. The complex rapidly dissociates unless the Arg(234)-Glu(446) salt bridge is released whereupon conformational changes occur that activate the metal ion-dependent adhesion site and partially stabilize the complex. The remaining salt bridge is then released, exposing the scissile bond and permitting factor D cleavage.


Asunto(s)
Complemento C3b/química , Factor B del Complemento/química , Factor D del Complemento/química , Complejos Multienzimáticos/química , Sustitución de Aminoácidos , Complemento C3b/genética , Complemento C3b/metabolismo , Factor B del Complemento/genética , Factor B del Complemento/metabolismo , Factor D del Complemento/genética , Factor D del Complemento/metabolismo , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Mutación Missense , Unión Proteica
8.
J Biol Chem ; 286(1): 123-30, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21047788

RESUMEN

A wide variety of nanomaterials are currently being developed for use in the detection and treatment of human diseases. However, there is no systematic way to measure and predict the action of such materials in biological contexts. Lipid-encapsulated nanoparticles (NPs) are a class of nanomaterials that includes the liposomes, the most widely used and clinically proven type of NPs. Liposomes can, however, activate the complement system, an important branch of innate immunity, resulting in undesirable consequences. Here, we describe the complement response to lipid-encapsulated NPs that are functionalized on the surface with various lipid-anchored gadolinium chelates. We developed a quantitative approach to examine the interaction of NPs with the complement system using in vitro assays and correlating these results with those obtained in an in vivo mouse model. Our results indicate that surface functionalization of NPs with certain chemical structures elicits swift complement activation that is initiated by a natural IgM antibody and propagated via the classical pathway. The intensity of the response is dependent on the chemical structures of the lipid-anchored chelates and not zeta potential effects alone. Moreover, the extent of complement activation may be tempered by complement inhibiting regulatory proteins that bind to the surface of NPs. These findings represent a step forward in the understanding of the interactions between nanomaterials and the host innate immune response and provide the basis for a systematic structure-activity relationship study to establish guidelines that are critical to the future development of biocompatible nanotherapeutics.


Asunto(s)
Anticuerpos/inmunología , Proteínas del Sistema Complemento/inmunología , Nanocápsulas/química , Fosfolípidos , Animales , Diseño de Fármacos , Gadolinio/química , Humanos , Inmunoglobulina M/inmunología , Ratones , Nanocápsulas/efectos adversos , Propiedades de Superficie
9.
Proc Natl Acad Sci U S A ; 105(26): 9023-8, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18579773

RESUMEN

Apoptotic cells must be rapidly eliminated to avoid harmful inflammatory and autoimmune reactions. Innate immunity is designed/poised to identify dying cells by their unique surface-associated molecular patterns. Here we demonstrate for the first time, to our knowledge, that the human complement protein properdin binds to early apoptotic T cells and initiates complement activation, leading to C3b opsonization and ingestion by phagocytic cells. Properdin binding was facilitated by the glycosaminoglycan chains of surface proteoglycans. Properdin released by activated neutrophils was particularly effective at recognition of apoptotic T cells, whereas the binding activity of properdin in the serum appeared to be inhibited. "Properdin tagging" of apoptotic T cells also induced their uptake by phagocytes independent of complement activation or other complement proteins. Although our findings were made primarily with apoptotic T cells, they suggest that properdin could play a similar role during apoptosis of other cell types.


Asunto(s)
Apoptosis , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Activación de Complemento/inmunología , Fagocitosis/inmunología , Properdina/inmunología , Linfocitos T CD4-Positivos/patología , Complemento C3b/inmunología , Células Dendríticas/inmunología , Glicosaminoglicanos/inmunología , Humanos , Macrófagos/inmunología , Neutrófilos/inmunología , Fagocitos/citología , Fagocitos/inmunología , Unión Proteica , Proteoglicanos/inmunología
10.
J Immunol ; 179(4): 2600-8, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17675523

RESUMEN

Complement promotes the rapid recognition and elimination of pathogens, infected cells, and immune complexes. The biochemical basis for its target specificity is incompletely understood. In this report, we demonstrate that properdin can directly bind to microbial targets and provide a platform for the in situ assembly and function of the alternative pathway C3 convertases. This mechanism differs from the standard model wherein nascent C3b generated in the fluid phase attaches nonspecifically to its targets. Properdin-directed complement activation occurred on yeast cell walls (zymosan) and Neisseria gonorrhoeae. Properdin did not bind wild-type Escherichia coli, but it readily bound E. coli LPS mutants, and the properdin-binding capacity of each strain correlated with its respective serum-dependent AP activation rate. Moreover, properdin:single-chain Ab constructs were used to direct serum-dependent complement activation to novel targets. We conclude properdin participates in two distinct complement activation pathways: one that occurs by the standard model and one that proceeds by the properdin-directed model. The properdin-directed model is consistent with a proposal made by Pillemer and his colleagues >50 years ago.


Asunto(s)
C3 Convertasa de la Vía Alternativa del Complemento/química , Vía Alternativa del Complemento , Escherichia coli K12/química , Lipopolisacáridos/química , Neisseria gonorrhoeae/química , Properdina/química , Zimosan/química , Animales , Anticuerpos/química , Anticuerpos/genética , Anticuerpos/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Gonorrea/metabolismo , Humanos , Lipopolisacáridos/metabolismo , Mutación , Neisseria gonorrhoeae/metabolismo , Properdina/genética , Properdina/metabolismo , Unión Proteica , Conejos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ovinos , Células U937 , Zimosan/metabolismo
11.
Mol Immunol ; 42(1): 87-97, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15488947

RESUMEN

Studies in gene-targeted mice have demonstrated that factor B of the alternative complement pathway plays an important role in several disease models, but an exogenous inhibitor of factor B has not previously been available. We have developed an inhibitory monoclonal antibody directed against a critical epitope on mouse factor B and have tested it in a model of antiphospholipid (aPL) antibody (Ab)-induced fetal loss. Gene-targeted factor B-deficient mice (fB-/-) were injected with a fusion protein comprised of the second and third short consensus repeat (SCR) domains of mouse factor B linked to a mouse IgG1 Fc domain. Hybridomas were made from splenocytes of the immunized mouse. One mAb, designated 1379, produced an IgG1 antibody that inhibited alternative pathway activation in vitro and in vivo by preventing formation of the C3bBb complex. Strikingly, this mAb inhibited alternative pathway activation in serum from mice, rats, humans, monkeys, pigs and horses. Fab fragments made from this mAb also inhibited alternative pathway activation. Epitope mapping demonstrated that this antibody binds to factor B within the third SCR domain. When mAb 1379 was administered to mice that also received human IgG containing antiphospholipid antibodies, it provided significant protection from antiphospholipid antibody-induced complement activation and fetal loss. Thus, this mAb to factor B has broad species reactivity and effectively inhibits alternative pathway activation. The mAb protects mice in an in vivo model of antiphospholipid antibody syndrome, demonstrating the therapeutic potential for the inhibition of factor B in this disease.


Asunto(s)
Anticuerpos Antifosfolípidos/efectos adversos , Activación de Complemento/efectos de los fármacos , Muerte Fetal/prevención & control , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Factor B del Complemento/antagonistas & inhibidores , Factor B del Complemento/inmunología , Modelos Animales de Enfermedad , Mapeo Epitopo , Femenino , Muerte Fetal/inmunología , Humanos , Ratones , Ratones Noqueados , Embarazo , Complicaciones Hematológicas del Embarazo/tratamiento farmacológico
12.
J Biol Chem ; 278(52): 52386-91, 2003 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-14561755

RESUMEN

The cleavage of C3 by the C3 convertases (C3bBb and C4b2a) determines whether complement activation proceeds. Dissociation (decay acceleration) of these central enzymes by the regulators decay-accelerating factor (DAF), complement receptor 1 (CR1), factor H, and C4-binding protein (C4BP) controls their function. In a previous investigation, we obtained evidence implicating the alpha4/5 region of the type A domain of Bb (especially Tyr338) in decay acceleration of C3bBb and proposed this site as a potential interaction point with DAF and long homologous repeat A of CR1. Because portions of only two DAF complement control protein domains (CCPs), CCP2 and CCP3, are necessary to mediate its decay of the CP C3 convertase (as opposed to portions of at least three CCPs in all other cases, e.g. CCPs 1-3 of CR1), DAF/C4b2a provides the simplest structural model for this reaction. Therefore, we examined the importance of the C2 alpha4/5 site on decay acceleration of C4b2a. Functional C4b2a complexes made with the C2 Y327A mutant, the C2 homolog to factor B Y338A, were highly resistant to DAF, C4BP, and long homologous repeat A of CR1, whereas C2 substitutions in two nearby residues (N324A and L328A) resulted in partial resistance. Our new findings indicate that the alpha4/5 region of C2a is critical to decay acceleration mediated by DAF, C4BP, and CR1 and suggest that decay acceleration of C4b2a and C3bBb requires interaction of the convertase alpha4/5 region with a CCP2/CCP3 site of DAF or structurally homologous sites of CR1 and C4BP.


Asunto(s)
Convertasas de Complemento C3-C5/química , Proteínas Inactivadoras de Complemento , Tirosina/química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Convertasas de Complemento C3-C5/metabolismo , Relación Dosis-Respuesta a Droga , Glicoproteínas/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Receptores de Complemento 3b/química , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...