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










Base de dados
Intervalo de ano de publicação
2.
J Immunol ; 184(11): 6256-65, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20435933

RESUMO

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.


Assuntos
Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico/genética , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Linfócitos B/imunologia , Separação Celular , Citometria de Fluxo , Imunofluorescência , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Congênicos , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia
4.
Arthritis Rheum ; 58(7): 2131-41, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18576325

RESUMO

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.


Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Cromossomos Humanos Par 1/imunologia , Switching de Imunoglobulina/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Autoanticorpos , DNA de Cadeia Simples , Humanos , Imunoglobulina G , Imunoglobulina M , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo
5.
J Clin Invest ; 118(2): 608-18, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202746

RESUMO

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.


Assuntos
Complemento C3/metabolismo , Fator H do Complemento/deficiência , Fator I do Complemento/fisiologia , Glomerulonefrite Membranoproliferativa/genética , Animais , Complemento C3/análise , Fator H do Complemento/administração & dosagem , Fator H do Complemento/genética , Fator I do Complemento/administração & dosagem , Fator I do Complemento/genética , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/imunologia , Camundongos , Camundongos Mutantes
6.
Mol Immunol ; 45(3): 787-95, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17675234

RESUMO

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.


Assuntos
Anticorpos Antinucleares/imunologia , Complemento C1q/deficiência , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Modelos Imunológicos , Tolerância a Antígenos Próprios , Transgenes/imunologia , Animais , Anticorpos Antinucleares/genética , Morte Celular/genética , Morte Celular/imunologia , Centro Germinativo/imunologia , Centro Germinativo/patologia , Imunoglobulina G/genética , Imunoglobulina M/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Transgênicos , Plasmócitos/imunologia , Plasmócitos/patologia , Tolerância a Antígenos Próprios/genética
7.
J Exp Med ; 204(6): 1249-56, 2007 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-17517971

RESUMO

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.


Assuntos
Fator H do Complemento/genética , Glomerulonefrite Membranoproliferativa/genética , Síndrome Hemolítico-Urêmica/genética , Degeneração Macular/genética , Idoso , Animais , Western Blotting , Complemento C3/metabolismo , Primers do DNA , Olho/patologia , Haplótipos/genética , Síndrome Hemolítico-Urêmica/patologia , Humanos , Rim/patologia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Estrutura Terciária de Proteína
8.
J Immunol ; 178(4): 2352-60, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17277141

RESUMO

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.


Assuntos
Formação de Anticorpos/genética , Autoimunidade/genética , Cromossomos/genética , Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico/genética , Locos de Características Quantitativas/genética , Animais , Anticorpos Antinucleares/imunologia , Formação de Anticorpos/imunologia , Autoantígenos/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Cromossomos/imunologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Locos de Características Quantitativas/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
9.
J Immunol ; 178(5): 2916-22, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17312136

RESUMO

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.


Assuntos
Autoantígenos/imunologia , Linfócitos B/imunologia , Complemento C1q/deficiência , Tolerância Imunológica , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Animais , Autoanticorpos/imunologia , Autoantígenos/genética , Linfócitos B/patologia , Morte Celular/genética , Morte Celular/imunologia , Complemento C1q/imunologia , Tolerância Imunológica/genética , Imunoglobulina M/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Fagocitose/genética , Fagocitose/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia
10.
J Immunol ; 177(8): 5155-62, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17015700

RESUMO

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.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Antígenos/metabolismo , Linfócitos B/imunologia , Ativação do Complemento/imunologia , Animais , Linfócitos B/fisiologia , Complemento C3/metabolismo , Imunoglobulina M/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos B/imunologia
11.
Arthritis Res Ther ; 8(3): R68, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16606442

RESUMO

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.


Assuntos
Autoanticorpos/sangue , DNA de Cadeia Simples/imunologia , Histonas/imunologia , Imunoglobulina G/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Anticorpos Antinucleares/sangue , Cromatina/imunologia , Modelos Animais de Doenças , Imunoglobulina G/sangue , Lúpus Eritematoso Sistêmico/sangue , Camundongos
12.
Nat Rev Mol Cell Biol ; 6(4): 341-4, 2005 04.
Artigo em Inglês | MEDLINE | ID: mdl-15768046

RESUMO

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?


Assuntos
Pesquisa/economia , Previsões , Pesquisa/organização & administração , Pesquisa/tendências , Reino Unido
13.
Eur J Immunol ; 35(1): 252-60, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15597324

RESUMO

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.


Assuntos
Via Clássica do Complemento , Imunoglobulina M/metabolismo , Macrófagos/imunologia , Animais , Apoptose , Células da Medula Óssea/imunologia , Complemento C1q/deficiência , Complemento C1q/genética , Complemento C3/metabolismo , Imunidade Inata , Imunoglobulina M/deficiência , Imunoglobulina M/genética , Técnicas In Vitro , Camundongos , Camundongos Knockout , Fagocitose , Linfócitos T/citologia , Linfócitos T/imunologia
14.
Eur J Immunol ; 34(12): 3713-22, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15517607

RESUMO

C1q deficiency in both humans and mice is strongly associated with autoimmunity. We have previously shown that bone marrow transplantation (BMT) restored C1q levels in C1q-deficient (C1qa(-/-)) mice. Here, we studied the effect of BMT on autoimmunity in C1qa(-/-) mice. Following irradiation, young C1qa(-/-) or wild-type MRL/Mp mice received bone marrow cells (BMC) from strain-matched wild-type or C1qa(-/-) animals. C1q levels increased rapidly when C1qa(-/-) mice received BMC from wild-type mice. Conversely, they decreased slowly in wild-type mice transplanted with C1qa(-/-) BMC. C1qa(-/-) animals transplanted with C1qa(-/-) BMC demonstrated accelerated disease when compared with wild-type mice given wild-type BMC. In contrast, a significant delay in the development of autoantibodies and glomerulonephritis was observed in C1qa(-/-) mice reconstituted with wild-type BMC, and the impaired clearance of apoptotic cells, previously described in C1qa(-/-) mice, was rectified. Moreover, the autoimmune disease was accelerated in wild-type mice given C1qa(-/-) BMC compared to animals transplanted with wild-type cells. These results provide supporting evidence that BMT may be a therapeutic option in the treatment of autoimmunity associated with human C1q deficiency.


Assuntos
Doenças Autoimunes/terapia , Transplante de Medula Óssea , Complemento C1q/metabolismo , Animais , Doenças Autoimunes/imunologia , Complemento C1q/deficiência , Complemento C1q/imunologia , Camundongos , Fagocitose/imunologia , Fagocitose/fisiologia , Baço/metabolismo
15.
J Immunol ; 173(7): 4277-85, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383556

RESUMO

To dissect the individual effects of the four non-MHC, autosomal loci (Bxs1 to Bxs4) that contribute to SLE susceptibility in BXSB mice, we generated congenic lines from chromosome 1 on a C57BL/10.Y(BXSB) (B10.Yaa) background for the intervals (values in megabases (Mb)) Bxs1 (46.3-89.2 Mb), Bxs1/4 (20.0-65.9 Mb), Bxs1/2 (64.4-159.0 Mb), and Bxs2/3 (105.4-189.0 Mb). Glomerulonephritis, qualitatively similar to that seen in the parental BXSB strain, developed in three of these congenic strains. Early onset, severe disease was observed in B10.Yaa.BXSB-Bxs2/3 congenic mice and caused 50% mortality by 12 mo. In B10.Yaa.BXSB-Bxs1/4 mice disease progressed more slowly, resulting in 13% mortality at 12 mo. The progression of renal disease in both of these strains was correlated with the level of anti-dsDNA Abs. B10.Yaa.BXSB-Bxs1 mice, despite their genetic similarity to B10.Yaa.BXSB-Bxs1/4 mice, developed a low-grade glomerulonephritis in the absence of anti-dsDNA Abs. Thus, Bxs4 directed an increase in titer and spectrum of autoantibodies, whereas Bxs1 promoted the development of nephritis. The Bxs2 interval was linked to the production of anti-dsDNA Abs without concomitant glomerulonephritis. In contrast, the Bxs3 interval was sufficient to generate classic lupus nephritis in a nonautoimmune-prone strain. Immune phenotype differed between controls and congenics with a significant increase in B220(+) cells in BXSB and B10.Yaa.BXSB-Bxs2/3, and an increase in CD4 to CD8 ratio in both BXSB and B10.Yaa.BXSB-Bxs1/4. Disease in the Bxs3 mice was delayed in comparison to the BXSB parental strain, emphasizing the necessity for multiple interactions in the production of the full BXSB phenotype.


Assuntos
Mapeamento Cromossômico , Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Fenótipo , Animais , Autoanticorpos/biossíntese , Cruzamentos Genéticos , Feminino , Ligação Genética , Marcadores Genéticos/imunologia , Imunofenotipagem , Lúpus Eritematoso Sistêmico/mortalidade , Nefrite Lúpica/genética , Nefrite Lúpica/imunologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Índice de Gravidade de Doença , Esplenomegalia/genética , Esplenomegalia/imunologia
16.
Am J Pathol ; 165(3): 825-32, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331407

RESUMO

The terminal complement components C5a and the membrane attack complex are involved in the pathogenesis of ischemia-reperfusion injury in many organs. CD59 is the major regulator of membrane attack complex formation. Mice deficient in the Cd59a gene (mCd59a-/-) were used to investigate the role of CD59 in renal ischemia-reperfusion injury. Unilateral ischemia-reperfusion injury was induced by clamping the left renal pedicle for 30 minutes under general anesthetic. Mice were studied at 72 hours and 2 weeks after ischemia-reperfusion injury. mCd59a-/- mice developed significantly greater tubular injury (P = 0.01), tubulointerstitial apoptosis (P = 0.02), and neutrophil influx (P = 0.04) than controls at 72 hours after ischemia-reperfusion. Two weeks after ischemia-reperfusion, mCd59a-/- mice exhibited more severe tubular damage predominantly in a corticomedullary distribution than controls (P = 0.02). Quantification of interstitial leukocytes revealed significantly greater numbers of infiltrating lymphocytes (but not macrophages) in mCd59a-/- mice than controls (P = 0.04) at 2 weeks. At both time points, significantly more C9 (as a marker of membrane attack complex) deposition occurred in a peritubular distribution in mCd59a-/- mice than controls. In conclusion, these results demonstrate that the lack of CD59a, by allowing unregulated membrane attack complex deposition, exacerbates both the tubular injury and the interstitial leukocyte infiltrate after ischemia-reperfusion injury in mice.


Assuntos
Antígenos CD59/fisiologia , Complemento C9/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Túbulos Renais/lesões , Leucócitos/patologia , Traumatismo por Reperfusão/etiologia , Animais , Apoptose , Modelos Animais de Doenças , Homozigoto , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Leucócitos/metabolismo , Linfócitos/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
17.
PLoS Biol ; 2(8): E243, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15314659

RESUMO

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.


Assuntos
Autoimunidade/genética , Autoimunidade/imunologia , Modelos Genéticos , Animais , Núcleo Celular/metabolismo , Mapeamento Cromossômico , Estudos de Coortes , Modelos Animais de Doenças , Epistasia Genética , Feminino , Marcação de Genes , Ligação Genética , Predisposição Genética para Doença , Genoma , Genótipo , Lúpus Eritematoso Sistêmico/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Repetições de Microssatélites , Locos de Características Quantitativas , Especificidade da Espécie
18.
Immunology ; 113(1): 80-8, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15312138

RESUMO

C1q deficiency has been shown to accelerate spontaneous autoimmunity in mice. We studied the time course of activation of monocytes and lymphocytes in autoimmune and non-autoimmune mice in the presence or absence of C1q as a disease accelerator. Autoimmune MRL\Mp.C1qa-\- and non-autoimmune C57BL\6.C1qa-\- mice were analysed at various time points between 6 and 33 weeks of age and compared to strain- and age-matched C1q-sufficient controls. Splenic and peritoneal leucocytes were analysed by flow cytometry and plasma levels of immunoglobulin M (IgM), total IgG, IgG subclasses and IgM autoantibodies were measured. Both C1q-deficient strains had significantly more splenic monocytes than their controls at all time points analysed. In addition, MRL\Mp.C1qa-\- but not C57BL/6.C1qa-\- mice developed splenic hypercellularity starting at about 12-17 weeks old, had signs of accelerated CD4+ T-cell activation and showed a marked increase in splenic plasma cells and total serum IgM levels from about 22 weeks of age. The accelerated CD4+ T-cell activation was not due to a direct inhibitory effect of C1q on T cells. These data show that C1q deficiency causes splenic monocytosis together with accelerated T-cell activation in an autoimmune-prone mouse strain.


Assuntos
Doenças Autoimunes/imunologia , Complemento C1q/deficiência , Predisposição Genética para Doença , Ativação Linfocitária/imunologia , Monócitos/imunologia , Animais , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Divisão Celular/imunologia , Células Cultivadas , Feminino , Imunoglobulina M/sangue , Imunofenotipagem , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia
19.
Annu Rev Immunol ; 22: 431-56, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15032584

RESUMO

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.


Assuntos
Proteínas do Sistema Complemento/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Modelos Imunológicos , Transdução de Sinais/imunologia , Animais , Proteínas do Sistema Complemento/deficiência , Humanos , Lúpus Eritematoso Sistêmico/genética
20.
J Immunol ; 172(6): 3406-14, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15004139

RESUMO

Human CD93 (known as C1qRp) has been shown to be a phagocytic receptor involved in the in vitro C1q-dependent enhancement of phagocytosis. However, binding of CD93 to C1q and its function remain controversial. In this study, we have generated CD93-deficient mice (CD93(-/-)) to investigate its biological role(s). The CD93(-/-) mice were viable and showed no gross abnormalities in their development. Thioglycolate-elicited peritoneal macrophages deficient in CD93 showed a similar enhancement in complement- and FcgammaR-dependent uptake of RBC to the wild-type macrophages when plated on C1q-coated surfaces suggesting that the lack of this receptor had no effect on these C1q-mediated events. There was no impairment in either complement- or FcgammaR-dependent phagocytic assays in vivo. By contrast, the CD93(-/-) mice had a significant phagocytic defect in the clearance of apoptotic cells in vivo (human Jurkat T cells and murine thymocytes: p=0.0006 and p=0.0079, respectively) compared with strain-matched controls. However, in vitro, the CD93(-/-) macrophages showed similar engulfment of apoptotic cells to wild-type macrophages. Furthermore, no supporting evidence for a role of CD93 as an adhesion molecule was found using intravital microscopy or analyzing peritoneal cell recruitment in response to three different inflammatory stimuli (thioglycolate, zymosan A, and IL-1beta). Thus, our findings indicate that murine CD93 is expressed on the peritoneal macrophage, especially on thioglycolate-elicited cells, but does not appear to play a key role in C1q-mediated enhancement of phagocytosis or in the intercellular adhesion events tested. However, our results suggest that it may contribute to the in vivo clearance of dying cells.


Assuntos
Adjuvantes Imunológicos/fisiologia , Apoptose/imunologia , Complemento C1q/fisiologia , Receptores de Hialuronatos , Glicoproteínas de Membrana , Fagocitose/imunologia , Receptores de Complemento/fisiologia , Adjuvantes Imunológicos/deficiência , Adjuvantes Imunológicos/genética , Animais , Apoptose/genética , Proteínas de Transporte , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Complemento C3/metabolismo , Eritrócitos/imunologia , Eritrócitos/metabolismo , Marcação de Genes , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Células Jurkat , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais , Proteínas Opsonizantes/sangue , Proteínas Opsonizantes/metabolismo , Fagocitose/genética , Receptores de Complemento/biossíntese , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Receptores de IgG/sangue , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores de IgG/fisiologia , Deleção de Sequência/genética , Deleção de Sequência/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Tioglicolatos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...