Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Exp Med ; 204(6): 1249-56, 2007 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-17517971

RESUMEN

Factor H (FH) is an abundant serum glycoprotein that regulates the alternative pathway of complement-preventing uncontrolled plasma C3 activation and nonspecific damage to host tissues. Age-related macular degeneration (AMD), atypical hemolytic uremic syndrome (aHUS), and membranoproliferative glomerulonephritis type II (MPGN2) are associated with polymorphisms or mutations in the FH gene (Cfh), suggesting the existence of a genotype-phenotype relationship. Although AMD and MPGN2 share pathological similarities with the accumulation of complement-containing debris within the eye and kidney, respectively, aHUS is characterized by renal endothelial injury. This pathological distinction was reflected in our Cfh association analysis, which demonstrated that although AMD and MPGN2 share a Cfh at-risk haplotype, the haplotype for aHUS was unique. FH-deficient mice have uncontrolled plasma C3 activation and spontaneously develop MPGN2 but not aHUS. We show that these mice, transgenically expressing a mouse FH protein functionally equivalent to aHUS-associated human FH mutants, regulate C3 activation in plasma and spontaneously develop aHUS but not MPGN2. These animals represent the first model of aHUS and provide in vivo evidence that effective plasma C3 regulation and the defective control of complement activation on renal endothelium are the critical events in the molecular pathogenesis of FH-associated aHUS.


Asunto(s)
Factor H de Complemento/genética , Glomerulonefritis Membranoproliferativa/genética , Síndrome Hemolítico-Urémico/genética , Degeneración Macular/genética , Anciano , Animales , Western Blotting , Complemento C3/metabolismo , Cartilla de ADN , Ojo/patología , Haplotipos/genética , Síndrome Hemolítico-Urémico/patología , Humanos , Riñón/patología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Estructura Terciaria de Proteína
2.
Nat Genet ; 31(4): 424-8, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12091909

RESUMEN

The alternative pathway of complement is activated continuously in vivo through the C3 'tick-over' pathway. This pathway is triggered by the hydrolysis of C3, resulting in the formation of C3 convertase. This, in turn, generates C3b, which mediates many of the biological functions of complement. Factor H, the main regulator of this activation, prevents formation and promotes dissociation of the C3 convertase enzyme, and, together with factor I, mediates the proteolytic inactivation of C3b. Factor H deficiency, described in 29 individuals from 12 families and in pigs, allows unhindered activation of fluid-phase C3 and severe depletion of plasma C3 (ref. 11). Membranoproliferative glomerulonephritis (MPGN) occurs in factor H-deficient humans and pigs. Although MPGN has been reported in other conditions in which uncontrolled activation of C3 occurs, the role of C3 dysregulation in the pathogenesis of MPGN is not understood. Here we show that mice deficient in factor H (Cfh(-/-) mice) develop MPGN spontaneously and are hypersensitive to developing renal injury caused by immune complexes. Introducing a second mutation in the gene encoding complement factor B, which prevents C3 turnover in vivo, obviates the phenotype of Cfh(-/-) mice. Thus, uncontrolled C3 activation in vivo is essential for the development of MPGN associated with deficiency of factor H.


Asunto(s)
Activación de Complemento/genética , Complemento C3/inmunología , Factor H de Complemento/genética , Glomerulonefritis/genética , Glomerulonefritis/fisiopatología , Animales , Complemento C3/metabolismo , Complemento C9/metabolismo , Factor H de Complemento/metabolismo , Modelos Animales de Enfermedad , Femenino , Riñón/patología , Riñón/ultraestructura , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Mutación
3.
J Immunol ; 184(11): 6256-65, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20435933

RESUMEN

The 129-derived Sle16 is a susceptibility locus for systemic autoimmunity when present on the C57BL/6 (B6) background. Genetic analysis of a (129xB6)F2 cross identified a region from the B6 chromosome 3 (Sle18) with positive linkage to antinuclear Abs. In this study, we have generated a B6 congenic strain harboring the 129 allele of Sle18 and intercrossed this line with the lupus-prone B6.129-Sle16 strain. The presence of the 129-Sle18 allele in the B6.129-Sle16Sle18 double congenic mice suppressed the development of Sle16-mediated autoantibody production and ameliorated the renal pathology. The 129-Sle18 locus rectified the B cell abnormalities detected in the B6.129-Sle16 mice, such as the reduction in the percentage of marginal zone B and B1a cells and the increased number of germinal centers. The B6.129-Sle16Sle18 spleens still displayed an increased percentage of activated T and B cells. However, in the B6.129-Sle16Sle18 strain the percentage of naive T cells was equivalent to that in B6.129-Sle18 and B6 mice and these cells showed a reduced proliferative response to anti-CD3 stimulation compared with B6.129-Sle16 T cells. There was a significant increase in the percentage of CD4(+)FoxP3(+)regulatory T cells in all congenic strains. These cells had normal regulatory function when tested in vitro. Thus, 129-Sle18 represents a novel, non-MHC lupus-suppressor locus probably operating as a functional modifier of B cells that, in combination with other factors, leads to lupus resistance. Further characterization of this locus will help to uncover the immune mechanism(s) conferring protection against lupus.


Asunto(s)
Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/genética , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Congénicos , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
4.
J Clin Invest ; 118(2): 608-18, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18202746

RESUMEN

The inflammatory kidney disease membranoproliferative glomerulonephritis type II (MPGN2) is associated with dysregulation of the alternative pathway of complement activation. MPGN2 is characterized by the presence of complement C3 along the glomerular basement membrane (GBM). Spontaneous activation of C3 through the alternative pathway is regulated by 2 plasma proteins, factor H and factor I. Deficiency of either of these regulators results in uncontrolled C3 activation, although the breakdown of activated C3 is dependent on factor I. Deficiency of factor H, but not factor I, is associated with MPGN2 in humans, pigs, and mice. To explain this discordance, mice with single or combined deficiencies of these factors were studied. MPGN2 did not develop in mice with combined factor H and I deficiency or in mice deficient in factor I alone. However, administration of a source of factor I to mice with combined factor H and factor I deficiency triggered both activated C3 fragments in plasma and GBM C3 deposition. Mouse renal transplant studies demonstrated that C3 deposited along the GBM was derived from plasma. Together, these findings provide what we believe to be the first evidence that factor I-mediated generation of activated C3 fragments in the circulation is a critical determinant for the development of MPGN2 associated with factor H deficiency.


Asunto(s)
Complemento C3/metabolismo , Factor H de Complemento/deficiencia , Factor I de Complemento/fisiología , Glomerulonefritis Membranoproliferativa/genética , Animales , Complemento C3/análisis , Factor H de Complemento/administración & dosificación , Factor H de Complemento/genética , Factor I de Complemento/administración & dosificación , Factor I de Complemento/genética , Glomerulonefritis Membranoproliferativa/sangre , Glomerulonefritis Membranoproliferativa/inmunología , Ratones , Ratones Mutantes
6.
J Exp Med ; 195(6): 665-72, 2002 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-11901193

RESUMEN

By generating four IgG isotype-switch variants of the high affinity 34-3C anti-erythrocyte autoantibody, and comparing them to the IgG variants of the low affinity 4C8 anti-erythrocyte autoantibody that we have previously studied, we evaluated in this study how high affinity binding to erythrocytes influences the pathogenicity of each IgG isotype in relation to the respective contributions of Fcgamma receptor (FcgammaR) and complement. The 34-3C autoantibody opsonizing extensively circulating erythrocytes efficiently activated complement in vivo (IgG2a = IgG2b > IgG3), except for the IgG1 isotype, while the 4C8 IgG autoantibody failed to activate complement. The pathogenicity of the 34-3C autoantibody of IgG2b and IgG3 isotypes was dramatically higher (>200-fold) than that of the corresponding isotypes of the 4C8 antibody. This enhanced activity was highly (IgG2b) or totally (IgG3) dependent on complement. In contrast, erythrocyte-binding affinities only played a minor role in in vivo hemolytic activities of the IgG1 and IgG2a isotypes of 34-3C and 4C8 antibodies, where complement was not or only partially involved, respectively. The remarkably different capacities of four different IgG isotypes of low and high affinity anti-erythrocyte autoantibodies to activate FcgammaR-bearing effector cells and complement in vivo demonstrate the role of autoantibody affinity maturation and of IgG isotype switching in autoantibody-mediated pathology.


Asunto(s)
Autoanticuerpos/inmunología , Activación de Complemento/inmunología , Eritrocitos/inmunología , Inmunoglobulina G/inmunología , Anemia Hemolítica Autoinmune/etiología , Anemia Hemolítica Autoinmune/inmunología , Animales , Cambio de Clase de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos BALB C , Receptores de IgG/inmunología
7.
Mol Immunol ; 45(3): 787-95, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17675234

RESUMEN

C1q-deficient mice have been shown to develop a lupus-like disease and to display an impaired clearance of apoptotic cells that are enriched in lupus autoantigens. However, the role of C1q in the regulation of autoreactive B cells remains debatable. To explore this we crossed MRL/Mp C1q-deficient mice with knock-in transgenic (Tg) mice expressing an anti-ssDNA antibody (VH3H9R and VH3H9R/VLkappa8R). Analysis of the VH3H9R mice showed that in the absence of C1q higher titres of Tg-derived IgM and IgG3 anti-ssDNA antibodies were detectable. In contrast, in the VH3H9R/VLkappa8R C1q-deficient animals no increase in Tg antibody levels was observed. In both models the lack of C1q induced a marked reduction of marginal zone B cells and this was paralleled by a significant increase in the percentage of plasmocytes. Thus, one could postulate that in the absence of C1q the failure to clear efficiently dying cells provides an additional stimulus to the autoreactive Tg B cells resulting in their emigration from the marginal zone B cell compartment with subsequent increase in plasmocytes. However, the lack of C1q led to an increased production of Tg IgM and IgG3 antibodies only in VH3H9R mice indicating that additional genetic susceptibility factors are required to break self-tolerance.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Complemento C1q/deficiencia , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Modelos Inmunológicos , Autotolerancia , Transgenes/inmunología , Animales , Anticuerpos Antinucleares/genética , Muerte Celular/genética , Muerte Celular/inmunología , Centro Germinal/inmunología , Centro Germinal/patología , Inmunoglobulina G/genética , Inmunoglobulina M/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Transgénicos , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Autotolerancia/genética
8.
PLoS Biol ; 2(8): E243, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15314659

RESUMEN

Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder in which complex genetic factors play an important role. Several strains of gene-targeted mice have been reported to develop SLE, implicating the null genes in the causation of disease. However, hybrid strains between 129 and C57BL/6 mice, widely used in the generation of gene-targeted mice, develop spontaneous autoimmunity. Furthermore, the genetic background markedly influences the autoimmune phenotype of SLE in gene-targeted mice. This suggests an important role in the expression of autoimmunity of as-yet-uncharacterised background genes originating from these parental mouse strains. Using genome-wide linkage analysis, we identified several susceptibility loci, derived from 129 and C57BL/6 mice, mapped in the lupus-prone hybrid (129 x C57BL/6) model. By creating a C57BL/6 congenic strain carrying a 129-derived Chromosome 1 segment, we found that this 129 interval was sufficient to mediate the loss of tolerance to nuclear antigens, which had previously been attributed to a disrupted gene. These results demonstrate important epistatic modifiers of autoimmunity in 129 and C57BL/6 mouse strains, widely used in gene targeting. These background gene influences may account for some, or even all, of the autoimmune traits described in some gene-targeted models of SLE.


Asunto(s)
Autoinmunidad/genética , Autoinmunidad/inmunología , Modelos Genéticos , Animales , Núcleo Celular/metabolismo , Mapeo Cromosómico , Estudios de Cohortes , Modelos Animales de Enfermedad , Epistasis Genética , Femenino , Marcación de Gen , Ligamiento Genético , Predisposición Genética a la Enfermedad , Genoma , Genotipo , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Repeticiones de Microsatélite , Sitios de Carácter Cuantitativo , Especificidad de la Especie
10.
Immunobiology ; 205(4-5): 395-406, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12396002

RESUMEN

Deficiency of classical pathway complement components displays a hierarchical association with the development of systemic lupus erythematosus (SLE). Individuals with deficiency of C1q, the first component of the classical pathway of activation, have the highest prevalence of SLE and the most severe manifestations of the disease. However, complement is also implicated in the effector inflammatory phase of the autoimmune response that characterizes SLE. Complement proteins are deposited in inflamed tissues causing consumption of complement. In addition, autoantibodies to C1q develop as part of the autoantibody response. Understanding how C1q deficiency results in the autoimmune phenotype of SLE may provide valuable clues to the role of the complement system in the maintenance of immune tolerance. In this review firstly we discuss the relationship between C1q deficiency and/or consumption and lupus. Secondly, we consider the links between apoptosis and complement. Finally we review the lessons we have learned from a murine model of C1q deficiency discussing the experimental evidence in support of the hypothesis that C1q may critically influence the immune response to self-antigens contained within the surface blebs generated by apoptotic cells.


Asunto(s)
Apoptosis/inmunología , Autoinmunidad/inmunología , Complemento C1q/fisiología , Vía Clásica del Complemento/fisiología , Lupus Eritematoso Sistémico/inmunología , Animales , Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos , Activación de Complemento , Complemento C1q/deficiencia , Complemento C1q/inmunología , Humanos
11.
Arthritis Rheum ; 58(7): 2131-41, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18576325

RESUMEN

OBJECTIVE: Systemic lupus erythematosus is a multifactorial disease with a strong genetic component. Previous studies have shown that a 129-derived chromosome 1 interval (Sle16) on the C57BL/6 (B6) background is sufficient to induce humoral autoimmunity. The aim of the present study was to elucidate the mechanisms by which this locus contributes to the loss of peripheral tolerance. METHODS: Anti-single-stranded DNA (anti-ssDNA)-knockin transgenic mice (V(H)3H9R/Vkappa8R and V(H)3H9R) were crossed with a B6 congenic line named B6.129chr1b that carries the Sle16 locus. A parallel study of a gene-targeted animal, whose mutated gene is located within the 129chr1b interval on chromosome 1, was also performed. RESULTS: The combination of V(H)3H9R/Vkappa8R with the 129chr1b interval resulted in impaired B cell anergy, and transgenic IgM and IgG anti-ssDNA antibodies were found in the circulation. The presence of IgG2a(a) anti-ssDNA and IgM(a) anti-Sm antibodies in sera indicated that the autoreactive transgenic B cells underwent class switching and epitope spreading. The 129chr1b locus appeared to have a dominant effect, since transgenic antibodies were also detected in mice carrying a single allele. The gene-targeted animals showed a similar phenotype. CONCLUSION: The presence of a single 129chr1b locus on the B6 background impaired B cell anergy, prevented deletion of anti-DNA transgenic B cells, and induced receptor revision. The findings of this study also emphasize that the autoimmune phenotype observed in mice with targeted genes located on chromosome 1 may simply arise from epistatic interactions between the 129 and B6 parental strains.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Cromosomas Humanos Par 1/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Autoanticuerpos , ADN de Cadena Simple , Humanos , Inmunoglobulina G , Inmunoglobulina M , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo
12.
J Immunol ; 178(4): 2352-60, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17277141

RESUMEN

Extensive evidence indicates that genetic predisposition is a central element in susceptibility to systemic lupus erythematosus both in humans and animals. We have previously shown that a congenic line carrying a 129-derived chromosome 1 interval on the C57BL/6 background developed humoral autoimmunity. To further dissect the contribution to autoimmunity of this 129 interval, we have created six subcongenic strains carrying fractions of the original 129 region and analyzed their serological and cellular phenotypes. At 1 year of age the congenic strain carrying a 129 interval between the microsatellites D1Mit15 (87.9 cM) and D1Mit115 (99.7 cM) (B6.129chr1b) had high levels of autoantibodies, while all the other congenic lines were not significantly different from the C57BL/6 controls. The B6.129chr1b strain displayed only mild proliferative glomerulonephritis despite high levels of IgG and C3 deposited in the kidneys. FACS analysis of the spleens revealed that the B6.129chr1b mice had a marked increase in the percentage of activated T cells associated with a significant reduction in the proportion of CD4(+)CD25(high) regulatory T cells. Moreover, this analysis showed a significantly reduced percentage of marginal zone B cells that preceded autoantibody production. Interestingly the 129chr1b-expressing bone marrow-derived macrophages displayed an impaired uptake of apoptotic cells in vitro. Collectively, our data indicate that the 129chr1b segment when recombined on the C57BL/6 genomic background is sufficient to induce loss of tolerance to nuclear Ags. These findings have important implication for the interpretation of the autoimmune phenotype associated with gene-targeted models.


Asunto(s)
Formación de Anticuerpos/genética , Autoinmunidad/genética , Cromosomas/genética , Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/genética , Sitios de Carácter Cuantitativo/genética , Animales , Anticuerpos Antinucleares/inmunología , Formación de Anticuerpos/inmunología , Autoantígenos/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Cromosomas/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Lupus Eritematoso Sistémico/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Sitios de Carácter Cuantitativo/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
13.
J Immunol ; 178(5): 2916-22, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17312136

RESUMEN

Inherited deficiency of early components of the classical complement pathway is strongly associated with the targeting of intracellular self Ags in systemic lupus erythematosus, but the reasons for this association are debated. In this study, we show that C1q deficiency increases the positive selection of B1b B cells and IgM autoantibodies by an intracellular self Ag, which is exposed on dying cells, and decreases the negative selection of autoreactive conventional B cells by the same Ag. These effects are specific to intracellular Ag because C1q deficiency does not affect negative selection by extracellular self Ag or increase the positive selection of naive B cells. The B1-derived IgM autoantibody binds to the intracellular Ag when it is expressed on dying cells, leading to fixation of C1q and clearance of cells by phagocytosis. These findings suggest that the positive selection of autoreactive B1 cells by self Ags may contribute to the IgM and C1q-dependent clearance of dying cells in a feedback loop that limits exposure of conventional B cells to immunogenic self Ags. We show that exposure of intracellular Ag leads to the activation of conventional B cells, when there is a source of T cell help in vivo.


Asunto(s)
Autoantígenos/inmunología , Linfocitos B/inmunología , Complemento C1q/deficiencia , Tolerancia Inmunológica , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/genética , Linfocitos B/patología , Muerte Celular/genética , Muerte Celular/inmunología , Complemento C1q/inmunología , Tolerancia Inmunológica/genética , Inmunoglobulina M/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Fagocitosis/genética , Fagocitosis/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
14.
J Immunol ; 177(8): 5155-62, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17015700

RESUMEN

Coligation of CD21 with BCR on the surface of B cells provides a costimulatory signal essential for efficient Ab responses to T-dependent Ags. To achieve this, Ag must be directly linked to C3 fragments, but how this occurs in vivo is not fully understood. Using BCR transgenic mice, we demonstrated that C3 was deposited on the surface of B cells following both high- and moderate-affinity Ag binding. This was dependent on the specific binding of IgM to the BCR-bound Ag and can occur independently of soluble immune complex formation. Based on these data, we propose a novel model in which immune complexes can form directly on the surface of the B cell following Ag binding. This model has implications for our understanding of B lymphocyte activation.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Antígenos/metabolismo , Linfocitos B/inmunología , Activación de Complemento/inmunología , Animales , Linfocitos B/fisiología , Complemento C3/metabolismo , Inmunoglobulina M/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos B/inmunología
15.
Arthritis Res Ther ; 8(3): R68, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16606442

RESUMEN

Systemic lupus erythematosus (SLE) is characterised by the production of autoantibodies against ubiquitous antigens, especially nuclear components. Evidence makes it clear that the development of these autoantibodies is an antigen-driven process and that immune complexes involving DNA-containing antigens play a key role in the disease process. In rodents, DNase I is the major endonuclease present in saliva, urine and plasma, where it catalyses the hydrolysis of DNA, and impaired DNase function has been implicated in the pathogenesis of SLE. In this study we have evaluated the effects of transgenic over-expression of murine DNase I endonucleases in vivo in a mouse model of lupus. We generated transgenic mice having T-cells that express either wild-type DNase I (wt.DNase I) or a mutant DNase I (ash.DNase I), engineered for three new properties - resistance to inhibition by G-actin, resistance to inhibition by physiological saline and hyperactivity compared to wild type. By crossing these transgenic mice with a murine strain that develops SLE we found that, compared to control non-transgenic littermates or wt.DNase I transgenic mice, the ash.DNase I mutant provided significant protection from the development of anti-single-stranded DNA and anti-histone antibodies, but not of renal disease. In summary, this is the first study in vivo to directly test the effects of long-term increased expression of DNase I on the development of SLE. Our results are in line with previous reports on the possible clinical benefits of recombinant DNase I treatment in SLE, and extend them further to the use of engineered DNase I variants with increased activity and resistance to physiological inhibitors.


Asunto(s)
Autoanticuerpos/sangre , ADN de Cadena Simple/inmunología , Histonas/inmunología , Inmunoglobulina G/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Anticuerpos Antinucleares/sangre , Cromatina/inmunología , Modelos Animales de Enfermedad , Inmunoglobulina G/sangre , Lupus Eritematoso Sistémico/sangre , Ratones
16.
Nat Rev Mol Cell Biol ; 6(4): 341-4, 2005 04.
Artículo en Inglés | MEDLINE | ID: mdl-15768046

RESUMEN

In July 2004, the UK government published a 10-year science and innovation framework aimed at providing a more strategic, partnership-based approach to delivering science. With the aim of creating a UK society that is confident about the governance, regulation and use of science and technology, how can we sustain or increase the supply of money for research, how should funding agencies dispense money, and how can we optimize the partnership arrangements for the funding of research?


Asunto(s)
Investigación/economía , Predicción , Investigación/organización & administración , Investigación/tendencias , Reino Unido
17.
Eur J Immunol ; 35(1): 252-60, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15597324

RESUMEN

Soluble molecules including complement components have been shown to facilitate the clearance of dying cells by phagocytes, a process that is important in preventing tissue damage and autoimmunity. However, the extent to which complement is involved in this process and the relative contribution of each of the complement activation pathways is not fully understood. We examined the role of complement in the recognition/uptake of apoptotic thymocytes by murine bone marrow-derived macrophages (BMDM) in vitro using sera from gene-targeted mice. We found this process to be IgM- and complement-dependent, especially when the apoptotic cell-to-BMDM ratio was low, and the level of C3 deposition on apoptotic cells correlated closely with their uptake. The addition of C1q rectified the phagocytic defect seen in the presence of C1q-deficient serum in vitro but had no effect on the phagocytic defect observed with serum deficient in both IgM antibodies and C1q. Similarly, complement activation by IgM antibodies was essential for in vivo C3 deposition on apoptotic cells and their uptake by peritoneal macrophages. Hence, the efficient uptake of dying cells by BMDM requires IgM antibodies and complement.


Asunto(s)
Vía Clásica del Complemento , Inmunoglobulina M/metabolismo , Macrófagos/inmunología , Animales , Apoptosis , Células de la Médula Ósea/inmunología , Complemento C1q/deficiencia , Complemento C1q/genética , Complemento C3/metabolismo , Inmunidad Innata , Inmunoglobulina M/deficiencia , Inmunoglobulina M/genética , Técnicas In Vitro , Ratones , Ratones Noqueados , Fagocitosis , Linfocitos T/citología , Linfocitos T/inmunología
18.
Arthritis Res ; 4 Suppl 3: S279-93, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12110148

RESUMEN

Complement is implicated in the pathogenesis of systemic lupus erythematosus (SLE) in several ways and may act as both friend and foe. Homozygous deficiency of any of the proteins of the classical pathway is causally associated with susceptibility to the development of SLE, especially deficiency of the earliest proteins of the activation pathway. However, complement is also implicated in the effector inflammatory phase of the autoimmune response that characterizes the disease. Complement proteins are deposited in inflamed tissues and, in experimental models, inhibition of C5 ameliorates disease in a murine model. As a further twist to the associations between the complement system and SLE, autoantibodies to some complement proteins, especially to C1q, develop as part of the autoantibody response. The presence of anti-C1q autoantibodies is associated with severe illness, including glomerulonephritis. In this chapter the role of the complement system in SLE is reviewed and hypotheses are advanced to explain the complex relationships between complement and lupus.


Asunto(s)
Complemento C1q/inmunología , Lupus Eritematoso Sistémico/inmunología , Autoanticuerpos/inmunología , Humanos , Lupus Eritematoso Sistémico/etiología
19.
Annu Rev Immunol ; 22: 431-56, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15032584

RESUMEN

Complement has both beneficial and deleterious roles in the pathogenesis of systemic lupus erythematosus (SLE). On the one hand, patients with SLE present with decreased complement levels and with complement deposition in inflamed tissues, suggestive of a harmful role of complement in the effector phase of disease. On the other hand, homozygous deficiency of any of the classical pathway proteins is strongly associated with the development of SLE. There are two main hypotheses to explain these observations. The first invokes an important role for complement in the physiological waste-disposal mechanisms of dying cells and immune complexes. The second hypothesis is based around the role of complement in determining the activation thresholds of B and T lymphocytes, with the proposal that complement deficiency causes incomplete maintenance of peripheral tolerance. These two hypotheses are not mutually exclusive. In addition, there is evidence for a contribution from other genetic factors in determining the phenotype of disease in the absence of complement.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Lupus Eritematoso Sistémico/inmunología , Modelos Inmunológicos , Transducción de Señal/inmunología , Animales , Proteínas del Sistema Complemento/deficiencia , Humanos , Lupus Eritematoso Sistémico/genética
20.
J Immunol ; 170(6): 3223-32, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12626581

RESUMEN

Deficiency of complement in humans and mice is associated with the development of lupus and with abnormal repair of inflammatory and immune complex-mediated tissue injury. Here we ask whether similar defects in the resolution of inflammation are found in mice prone to spontaneous lupus. We compared the response to an i.p. injection of thioglycolate between two lupus-prone strains (MRL/Mp and NZB/W) and two non lupus-prone strains of mice (C57BL/6 and BALB/c). In all four strains the influx of polymorphonuclear neutrophils (PMN) was similar. However, by 96 h clearance of PMN in the control strains was complete, whereas in the autoimmune-prone strains PMN were still detectable. The number of mononuclear cells recruited was markedly reduced in the lupus-prone strains compared with the controls, and their phenotype was different. The lupus-prone strains had significantly fewer elicited macrophages that were CD11b-high and Ly6C-negative. In lupus-prone mice at 24 h there was a significantly increased number of apoptotic PMN free in the peritoneum, accompanied by a reduced percentage of macrophages containing apoptotic bodies, suggesting a defect in their uptake. An impaired ability of resident peritoneal macrophages from lupus-prone mice to engulf apoptotic cells was demonstrated by in vivo and in vitro cell clearance assays. These observations indicate that lupus-prone strains have an abnormal inflammatory response to thioglycolate and an intrinsic impairment in apoptotic cell uptake. These findings have implications for the initiation of autoimmunity, as lupus autoantigens are expressed on dying cells, and impaired disposal of these could enhance the development of autoimmunity.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Tioglicolatos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Eritrocitos/inmunología , Eritrocitos/metabolismo , Femenino , Inyecciones Intraperitoneales , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Ratones Endogámicos MRL lpr , Ratones Endogámicos NZB , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Proteínas Opsoninas/sangre , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/patología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA