Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Nat Immunol ; 18(7): 716-724, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28632714

RESUMO

Efforts to understand autoimmunity have been pursued relentlessly for several decades. It has become apparent that the immune system evolved multiple mechanisms for controlling self-reactivity, and defects in one or more of these mechanisms can lead to a breakdown of tolerance. Among the multitude of lesions associated with disease, the most common seem to affect peripheral tolerance rather than central tolerance. The initial trigger for both systemic autoimmune disorders and organ-specific autoimmune disorders probably involves the recognition of self or foreign molecules, especially nucleic acids, by innate sensors. Such recognition, in turn, triggers inflammatory responses and the engagement of previously quiescent autoreactive T cells and B cells. Here we summarize the most prominent autoimmune pathways and identify key issues that require resolution for full understanding of pathogenic autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Tolerância a Antígenos Próprios/imunologia , Linfócitos T/imunologia , Animais , Tolerância Central/imunologia , Humanos , Tolerância Periférica/imunologia
2.
Nat Chem Biol ; 2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38191941

RESUMO

SLC15A4 is an endolysosome-resident transporter linked with autoinflammation and autoimmunity. Specifically, SLC15A4 is critical for Toll-like receptors (TLRs) 7-9 as well as nucleotide-binding oligomerization domain-containing protein (NOD) signaling in several immune cell subsets. Notably, SLC15A4 is essential for the development of systemic lupus erythematosus in murine models and is associated with autoimmune conditions in humans. Despite its therapeutic potential, the availability of quality chemical probes targeting SLC15A4 functions is limited. In this study, we used an integrated chemical proteomics approach to develop a suite of chemical tools, including first-in-class functional inhibitors, for SLC15A4. We demonstrate that these inhibitors suppress SLC15A4-mediated endolysosomal TLR and NOD functions in a variety of human and mouse immune cells; we provide evidence of their ability to suppress inflammation in vivo and in clinical settings; and we provide insights into their mechanism of action. Our findings establish SLC15A4 as a druggable target for the treatment of autoimmune and autoinflammatory conditions.

3.
Proc Natl Acad Sci U S A ; 119(14): e2200544119, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35349343

RESUMO

A function-impairing mutation (feeble) or genomic deletion of SLC15A4 abolishes responses of nucleic acid­sensing endosomal toll-like receptors (TLRs) and significantly reduces disease in mouse models of lupus. Here, we demonstrate disease reduction in homozygous and even heterozygous Slc15a4 feeble mutant BXSB male mice with a Tlr7 gene duplication. In contrast to SLC15A4, a function-impairing mutation of SLC15A3 did not diminish type I interferon (IFN-I) production by TLR-activated plasmacytoid dendritic cells (pDCs), indicating divergence of function between these homologous SLC15 family members. Trafficking to endolysosomes and function of SLC15A4 were dependent on the Adaptor protein 3 (AP-3) complex. Importantly, SLC15A4 was required for trafficking and colocalization of nucleic acid­sensing TLRs and their ligands to endolysosomes and the formation of the LAMP2+VAMP3+ hybrid compartment in which IFN-I production is initiated. Collectively, these findings define mechanistic processes by which SLC15A4 controls endosomal TLR function and suggest that pharmacologic intervention to curtail the function of this transporter may be a means to treat lupus and other endosomal TLR-dependent diseases.


Assuntos
Ácidos Nucleicos , Animais , Endossomos/metabolismo , Ligantes , Lisossomos/metabolismo , Proteínas de Membrana Transportadoras/genética , Camundongos , Receptores Toll-Like/metabolismo
4.
Nat Immunol ; 11(4): 335-43, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20190759

RESUMO

Here we describe a previously unknown form of inherited immunodeficiency revealed by an N-ethyl-N-nitrosourea-induced mutation called elektra. Mice homozygous for this mutation showed enhanced susceptibility to bacterial and viral infection and diminished numbers of T cells and inflammatory monocytes that failed to proliferate after infection and died via the intrinsic apoptotic pathway in response to diverse proliferative stimuli. They also had a greater proportion of T cells poised to replicate DNA, and their T cells expressed a subset of activation markers, suggestive of a semi-activated state. We positionally ascribe the elektra phenotype to a mutation in the gene encoding Schlafen-2 (Slfn2). Our findings identify a physiological role for Slfn2 in the defense against pathogens through the regulation of quiescence in T cells and monocytes.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Leucócitos Mononucleares/imunologia , Linfócitos T/imunologia , Animais , Apoptose/imunologia , Sequência de Bases , Separação Celular , Citometria de Fluxo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Mutação , Fenótipo , Transdução de Sinais/imunologia
5.
Clin Immunol ; 205: 75-82, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31175964

RESUMO

Considerable evidence indicates that autoimmune disease expression depends on both genetic and environmental factors. Among potential environmental triggers, occupational airway exposure to crystalline silica and virus infections have been linked to lupus and other autoimmune diseases in both humans and mouse models. Here, we hypothesized that combined silica and virus exposures synergize and induce autoimmune manifestations more effectively than single exposure to either of these factors, particularly in individuals with low genetic predisposition. Accordingly, infection with the model murine pathogen lymphocytic choriomenigitis virus (LCMV) in early life, followed by airway exposure to crystalline silica in adult life, induced lupus-like autoantibodies to several nuclear self-antigens including chromatin, RNP and Sm, concurrent with kidney lesions, in non-autoimmune C57BL/6 (B6) mice. In contrast, given individually, LCMV or silica were largely ineffectual in this strain. These results support a multihit model of autoimmunity, where exposure to different environmental factors acting on distinct immunostimulatory pathways complements limited genetic predisposition and increases the risk of autoimmunity above a critical threshold.


Assuntos
Infecções por Arenaviridae/imunologia , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Rim/imunologia , Pulmão/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Vírus da Coriomeningite Linfocítica , Dióxido de Silício/toxicidade , Silicose/imunologia , Animais , Infecções por Arenaviridae/complicações , Doenças Autoimunes/etiologia , Doenças Autoimunes/patologia , Cromatina/imunologia , Doença Crônica , Interação Gene-Ambiente , Predisposição Genética para Doença , Rim/patologia , Pulmão/patologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ribonucleoproteínas/imunologia , Silicose/etiologia , Silicose/patologia
6.
J Immunol ; 199(11): 3739-3747, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29055005

RESUMO

Type I IFN and nucleic acid-sensing TLRs are both strongly implicated in the pathogenesis of lupus, with most patients expressing IFN-induced genes in peripheral blood cells and with TLRs promoting type I IFNs and autoreactive B cells. About a third of systemic lupus erythematosus patients, however, lack the IFN signature, suggesting the possibility of type I IFN-independent mechanisms. In this study, we examined the role of type I IFN and TLR trafficking and signaling in xenobiotic systemic mercury-induced autoimmunity (HgIA). Strikingly, autoantibody production in HgIA was not dependent on the type I IFN receptor even in NZB mice that require type I IFN signaling for spontaneous disease, but was dependent on the endosomal TLR transporter UNC93B1 and the endosomal proton transporter, solute carrier family 15, member 4. HgIA also required the adaptor protein-3 complex, which transports TLRs from the early endosome to the late endolysosomal compartments. Examination of TLR signaling pathways implicated the canonical NF-κB pathway and the proinflammatory cytokine IL-6 in autoantibody production, but not IFN regulatory factor 7. These findings identify HgIA as a novel type I IFN-independent model of systemic autoimmunity and implicate TLR-mediated NF-κB proinflammatory signaling from the late endocytic pathway compartments in autoantibody generation.


Assuntos
Doenças Autoimunes/imunologia , Endossomos/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Proteínas de Membrana Transportadoras/metabolismo , Receptores Toll-Like/metabolismo , Animais , Autoanticorpos/metabolismo , Doenças Autoimunes/induzido quimicamente , Autoimunidade , Células Cultivadas , Feminino , Humanos , Interferon Tipo I/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lisossomos/metabolismo , Masculino , Proteínas de Membrana Transportadoras/genética , Mercúrio , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Transporte Proteico , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Receptores Toll-Like/genética , Xenobióticos
7.
Nature ; 502(7471): 327-332, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24107995

RESUMO

Progressive phases of multiple sclerosis are associated with inhibited differentiation of the progenitor cell population that generates the mature oligodendrocytes required for remyelination and disease remission. To identify selective inducers of oligodendrocyte differentiation, we performed an image-based screen for myelin basic protein (MBP) expression using primary rat optic-nerve-derived progenitor cells. Here we show that among the most effective compounds identifed was benztropine, which significantly decreases clinical severity in the experimental autoimmune encephalomyelitis (EAE) model of relapsing-remitting multiple sclerosis when administered alone or in combination with approved immunosuppressive treatments for multiple sclerosis. Evidence from a cuprizone-induced model of demyelination, in vitro and in vivo T-cell assays and EAE adoptive transfer experiments indicated that the observed efficacy of this drug results directly from an enhancement of remyelination rather than immune suppression. Pharmacological studies indicate that benztropine functions by a mechanism that involves direct antagonism of M1 and/or M3 muscarinic receptors. These studies should facilitate the development of effective new therapies for the treatment of multiple sclerosis that complement established immunosuppressive approaches.


Assuntos
Benzotropina/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Modelos Biológicos , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Animais , Antiparkinsonianos/farmacologia , Antiparkinsonianos/uso terapêutico , Benzotropina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura , Cuprizona/farmacologia , Cuprizona/uso terapêutico , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/patologia , Feminino , Cloridrato de Fingolimode , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/patologia , Proteína Proteolipídica de Mielina/farmacologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Nervo Óptico/citologia , Propilenoglicóis/farmacologia , Propilenoglicóis/uso terapêutico , Ratos , Receptor Muscarínico M1/antagonistas & inibidores , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/antagonistas & inibidores , Receptor Muscarínico M3/metabolismo , Recidiva , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
8.
Proc Natl Acad Sci U S A ; 113(41): 11501-11506, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27663736

RESUMO

A variable region fusion strategy was used to generate an immunosuppressive antibody based on a novel "stalk-knob" structural motif in the ultralong complementary-determining region (CDR) of a bovine antibody. The potent Kv1.3 channel inhibitory peptides Moka1-toxin and Vm24-toxin were grafted into different CDRs of the humanized antibodies BVK and Synagis (Syn) using both ß-sheet and coiled-coil linkers. Structure-activity relationship efforts led to generation of the fusion protein Syn-Vm24-CDR3L, which demonstrated excellent selectivity and potency against effector human memory T cells (subnanomolar to picomolar EC50 values). This fusion antibody also had significantly improved plasma half-life and serum stability in rodents compared with the parent Vm24 peptide. Finally, this fusion protein showed potent in vivo efficacy in the delayed type hypersensitivity in rats. These results illustrate the utility of antibody CDR fusions as a general and effective strategy to generate long-acting functional antibodies, and may lead to a selective immunosuppressive antibody for the treatment of autoimmune diseases.


Assuntos
Anticorpos Bloqueadores/farmacologia , Desenho de Fármacos , Imunossupressores/farmacologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Células CHO , Bovinos , Regiões Determinantes de Complementaridade/química , Cricetinae , Cricetulus , Células HEK293 , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia
9.
FASEB J ; 30(2): 738-47, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26481307

RESUMO

We have devised a method of using intracellular combinatorial libraries to select antibodies that control cell fates. Many agonist antibodies have been selected with this method, and the process appears to be limited only by the availability of a phenotypic selection system. We demonstrate the utility of this approach to discover agonist antibodies that engage an unanticipated target and regulate macrophage polarization by selective induction of anti-inflammatory M2 macrophages. This antibody was used therapeutically to block autoimmunity in a classic mouse model of spontaneous systemic lupus erythematosus.


Assuntos
Lúpus Eritematoso Cutâneo/imunologia , Macrófagos/imunologia , Anticorpos de Cadeia Única/farmacologia , Animais , Modelos Animais de Doenças , Células HEK293 , Humanos , Lúpus Eritematoso Cutâneo/tratamento farmacológico , Lúpus Eritematoso Cutâneo/patologia , Macrófagos/patologia , Camundongos , Anticorpos de Cadeia Única/imunologia
10.
Proc Natl Acad Sci U S A ; 111(24): 8925-30, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24889626

RESUMO

The outcome of a viral infection reflects the balance between virus virulence and host susceptibility. The clone 13 (Cl13) variant of lymphocytic choriomeningitis virus--a prototype of Old World arenaviruses closely related to Lassa fever virus--elicits in C57BL/6 and BALB/c mice abundant negative immunoregulatory molecules, associated with T-cell exhaustion, negligible T-cell-mediated injury, and high virus titers that persist. Conversely, here we report that in NZB mice, despite the efficient induction of immunoregulatory molecules and high viremia, Cl13 generated a robust cytotoxic T-cell response, resulting in thrombocytopenia, pulmonary endothelial cell loss, vascular leakage, and death within 6-8 d. These pathogenic events required type I IFN (IFN-I) signaling on nonhematopoietic cells and were completely abrogated by IFN-I receptor blockade. Thus, IFN-I may play a prominent role in hemorrhagic fevers and other acute virus infections associated with severe vascular pathology, and targeting IFN-I or downstream effector molecules may be an effective therapeutic approach.


Assuntos
Interferon Tipo I/metabolismo , Febre Lassa/virologia , Doenças Vasculares/virologia , Animais , Lavagem Broncoalveolar , Linhagem Celular , Cricetinae , Citocinas/metabolismo , Feminino , Vírus Lassa , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Transgênicos , Transdução de Sinais , Células-Tronco/química , Linfócitos T Citotóxicos/virologia , Ativação Viral
11.
Proc Natl Acad Sci U S A ; 110(8): 2940-5, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382217

RESUMO

In vitro evidence suggests that plasmacytoid dendritic cells (pDCs) are intimately involved in the pathogenesis of lupus. However, it remains to be determined whether these cells are required in vivo for disease development, and whether their contribution is restricted to hyperproduction of type I IFNs. To address these issues, we created lupus-predisposed mice lacking the IFN regulatory factor 8 (IRF8) or carrying a mutation that impairs the peptide/histidine transporter solute carrier family 15, member 4 (SLC15A4). IRF8-deficient NZB mice, lacking pDCs, showed almost complete absence of anti-nuclear, anti-chromatin, and anti-erythrocyte autoantibodies, along with reduced kidney disease. These effects were observed despite normal B-cell responses to Toll-like receptor (TLR) 7 and TLR9 stimuli and intact humoral responses to conventional T-dependent and -independent antigens. Moreover, Slc15a4 mutant C57BL/6-Fas(lpr) mice, in which pDCs are present but unable to produce type I IFNs in response to endosomal TLR ligands, also showed an absence of autoantibodies, reduced lymphadenopathy and splenomegaly, and extended survival. Taken together, our results demonstrate that pDCs and the production of type I IFNs by these cells are critical contributors to the pathogenesis of lupus-like autoimmunity in these models. Thus, IRF8 and SLC15A4 may provide important targets for therapeutic intervention in human lupus.


Assuntos
Células Dendríticas/imunologia , Fatores Reguladores de Interferon/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas de Membrana Transportadoras/fisiologia , Animais , Fatores Reguladores de Interferon/genética , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Clin Immunol ; 161(2): 260-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26319414

RESUMO

IL-7 is known to be vital for T cell homeostasis but has previously been presumed to be dispensable for TCR-induced activation. Here, we show that IL-7 is critical for the initial activation of CD4(+) T cells in that it provides some of the necessary early signaling components, such as activated STAT5 and Akt. Accordingly, short-term in vivo IL-7Rα blockade inhibited the activation and expansion of autoantigen-specific CD4(+) T cells and, when used to treat experimental autoimmune encephalomyelitis (EAE), prevented and ameliorated disease. Our studies demonstrate that IL-7 signaling is a prerequisite for optimal CD4(+) T cell activation and that IL-7R antagonism may be effective in treating CD4(+) T cell-mediated neuroinflammation and other autoimmune inflammatory conditions.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interleucina-7/imunologia , Ativação Linfocitária/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células , Citocinas/imunologia , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/metabolismo , Citometria de Fluxo , Humanos , Interleucina-7/deficiência , Interleucina-7/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Fosforilação/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Interleucina-7/imunologia , Receptores de Interleucina-7/metabolismo , Fator de Transcrição STAT2/imunologia , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais/imunologia
13.
J Immunol ; 190(10): 4982-90, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23589617

RESUMO

Nucleic acid (NA)-sensing TLRs (NA-TLRs) promote the induction of anti-nuclear Abs in systemic lupus erythematosus. However, the extent to which other nonnuclear pathogenic autoantibody specificities that occur in lupus and independently in other autoimmune diseases depend on NA-TLRs, and which immune cells require NA-TLRs in systemic autoimmunity, remains to be determined. Using Unc93b1(3d) lupus-prone mice that lack NA-TLR signaling, we found that all pathogenic nonnuclear autoantibody specificities examined, even anti-RBC, required NA-TLRs. Furthermore, we document that NA-TLRs in B cells were required for the development of antichromatin and rheumatoid factor. These findings support a unifying NA-TLR-mediated mechanism of autoantibody production that has both pathophysiological and therapeutic implications for systemic lupus erythematosus and several other humoral-mediated autoimmune diseases. In particular, our findings suggest that targeting of NA-TLR signaling in B cells alone would be sufficient to specifically block production of a broad diversity of autoantibodies.


Assuntos
Anticorpos Antinucleares/imunologia , Linfócitos B/imunologia , Glicoproteínas de Membrana/imunologia , Ácidos Nucleicos/imunologia , Receptor 7 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Animais , Células Produtoras de Anticorpos/imunologia , Autoanticorpos/imunologia , Células da Medula Óssea/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Cromatina/imunologia , Células Dendríticas , Feminino , Síndromes de Imunodeficiência , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Proteínas de Membrana Transportadoras/imunologia , Camundongos , Camundongos Endogâmicos NZB , Fator 88 de Diferenciação Mieloide/imunologia , Doenças da Imunodeficiência Primária , Fator Reumatoide/imunologia , Ribonucleoproteínas/imunologia , Transdução de Sinais
14.
Proc Natl Acad Sci U S A ; 109(31): 12650-5, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-22802641

RESUMO

Using an environmentally sensitized genetic screen we identified mutations that cause inflammatory colitis in mice. The X-linked Klein-Zschocher (KLZ) mutation created a null allele of Yipf6, a member of a gene family believed to regulate vesicular transport in yeast, but without known functions in mammals. Yipf6 is a five transmembrane-spanning protein associated with Golgi compartments. Klein-Zschocher mutants were extremely sensitive to colitis induced by dextran sodium sulfate (DSS) and developed spontaneous ileitis and colitis after 16 mo of age in specific pathogen-free housing conditions. Electron microscopy, gene expression, and immunocytochemistry analyses provided evidence that impaired intestinal homeostasis stemmed from defective formation and secretion of large secretory granules from Paneth and goblet cells. These studies support a tissue- and organ-specific function for Yipf6 in the maintenance of intestinal homeostasis and implicate the orthologous human gene as a disease susceptibility locus.


Assuntos
Colite/metabolismo , Células Caliciformes/metabolismo , Proteínas de Membrana/metabolismo , Mutação , Celulas de Paneth/metabolismo , Animais , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Sulfato de Dextrana/toxicidade , Feminino , Regulação da Expressão Gênica , Loci Gênicos , Predisposição Genética para Doença , Células Caliciformes/ultraestrutura , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Humanos , Ileíte/induzido quimicamente , Ileíte/genética , Ileíte/metabolismo , Ileíte/patologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Celulas de Paneth/ultraestrutura
15.
J Immunol ; 188(7): 3307-14, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22371393

RESUMO

The lupus-prone New Zealand Black (NZB) strain uniquely develops a genetically imposed severe spontaneous autoimmune hemolytic anemia (AIHA) that is very similar to the corresponding human disease. Previous studies have mapped anti-erythrocyte Ab (AEA)-promoting NZB loci to several chromosomal locations, including chromosome 4; however, none of these have been analyzed with interval congenics. In this study, we used NZB.NZW-Lbw2 congenic (designated Lbw2 congenic) mice containing an introgressed fragment of New Zealand White (NZW) on chromosome 4 encompassing Lbw2, a locus previously linked to survival, glomerulonephritis, and splenomegaly, to investigate its role in AIHA. Lbw2 congenic mice exhibited marked reductions in AEAs and splenomegaly but not in anti-nuclear Abs. Furthermore, Lbw2 congenics had greater numbers of marginal zone B cells and reduced expansion of peritoneal cells, particularly the B-1a cell subset at early ages, but no reduction in B cell response to LPS. Analysis of a panel of subinterval congenic mice showed that the full effect of Lbw2 on AEA production was dependent on three subloci, with splenomegaly mapping to two of the subloci and expansions of peritoneal cell populations, including B-1a cells to one. These results directly demonstrated the presence of AEA-specific promoting genes on NZB chromosome 4, documented a marked influence of background genes on autoimmune phenotypes related to Lbw2, and further refined the locations of the underlying genetic variants. Delineation of the Lbw2 genes should yield new insights into both the pathogenesis of AIHA and the nature of epistatic interactions of lupus-modifying genetic variants.


Assuntos
Anemia Hemolítica Autoimune/genética , Anemia Hemolítica Congênita/genética , Lúpus Eritematoso Sistêmico/genética , Camundongos Endogâmicos NZB/genética , Anemia Hemolítica Autoimune/imunologia , Anemia Hemolítica Autoimune/patologia , Anemia Hemolítica Congênita/imunologia , Anemia Hemolítica Congênita/patologia , Animais , Líquido Ascítico/imunologia , Líquido Ascítico/patologia , Autoanticorpos/biossíntese , Autoanticorpos/genética , Subpopulações de Linfócitos B/imunologia , Mapeamento Cromossômico , Teste de Coombs , Cruzamentos Genéticos , Eritrócitos/imunologia , Estudos de Associação Genética , Predisposição Genética para Doença , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos NZB/imunologia , Camundongos Endogâmicos , Baço/imunologia , Baço/patologia , Esplenomegalia/etiologia
16.
J Immunol ; 189(12): 5976-84, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23175700

RESUMO

The demonstration in humans and mice that nucleic acid-sensing TLRs and type I IFNs are essential disease mediators is a milestone in delineating the mechanisms of lupus pathogenesis. In this study, we show that Ifnb gene deletion does not modify disease progression in NZB mice, thereby strongly implicating IFN-α subtypes as the principal pathogenic effectors. We further document that long-term treatment of male BXSB mice with an anti-IFN-α/ß receptor Ab of mouse origin reduced serologic, cellular, and histologic disease manifestations and extended survival, suggesting that disease acceleration by the Tlr7 gene duplication in this model is mediated by type I IFN signaling. The efficacy of this treatment in BXSB mice was clearly evident when applied early in the disease process, but only partial reductions in some disease characteristics were observed when treatment was initiated at later stages. A transient therapeutic effect was also noted in the MRL-Fas(lpr) model, although overall mortality was unaffected. The combined findings suggest that IFN-α/ß receptor blockade, particularly when started at early disease stages, may be a useful treatment approach for human systemic lupus erythematosus and other autoimmune syndromes.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Autoanticorpos/administração & dosagem , Nefrite Lúpica/imunologia , Nefrite Lúpica/terapia , Receptor de Interferon alfa e beta/imunologia , Animais , Anticorpos Antinucleares/administração & dosagem , Anticorpos Antinucleares/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Autoanticorpos/biossíntese , Autoanticorpos/uso terapêutico , Células Cultivadas , Predisposição Genética para Doença/etiologia , Humanos , Nefrite Lúpica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Endogâmicos NZB , Camundongos Knockout , Proteinúria/genética , Proteinúria/imunologia , Proteinúria/terapia
17.
Nat Med ; 13(5): 543-51, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17479100

RESUMO

We formulate a two-phase paradigm of autoimmunity associated with systemic lupus erythematosus, the archetypal autoimmune disease. The initial Toll-like receptor (TLR)-independent phase is mediated by dendritic cell uptake of apoptotic cell debris and associated nucleic acids, whereas the subsequent TLR-dependent phase serves an amplification function and is mediated by uptake of TLR ligands derived from self-antigens (principally nucleic acids) complexed with autoantibodies. Both phases depend on elaboration of type I interferons (IFNs), and therapeutic interruption of induction or activity of these cytokines in predisposed individuals might have a substantial mitigating effect in lupus and other autoimmune diseases.


Assuntos
Autoimunidade/fisiologia , Interferon Tipo I/imunologia , Receptores Toll-Like/imunologia , Doenças Autoimunes/fisiopatologia , Humanos , Interferon-alfa/imunologia , Interferon beta/imunologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Modelos Imunológicos
18.
Ocul Surf ; 32: 130-138, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38395195

RESUMO

The literature is filled with citations reporting an increased incidence of chronic dry eye disease, also known as keratoconjunctivitis sicca, in patients with systemic autoimmune diseases such as rheumatoid arthritis, Sjögren's Syndrome, systemic sclerosis and lupus. As the most environmentally exposed mucosal surface of the body, the conjunctiva constantly responds to environmental challenges which are typically self limited, but when persistent and unresolved may provoke pathogenic innate and adaptive immune reactions. Our understanding of the pathophysiological mechanisms by which systemic autoimmune diseases cause dry eye inducing ocular surface inflammation continues to evolve. Conjunctival immune tone responds to self or foreign danger signals (including desiccating stress) on the ocular surface with an initial non-specific innate inflammatory response. If unchecked, this can lead to activation of dendritic cells that present antigen and prime T and B cells resulting in an adaptive immune reaction. These reactions generally resolve, but dysfunctional, hyper-responsive immune cells found in systemic autoimmune diseases that are recruited to the ocular surface can amplify inflammatory stress responses in the ocular surface and glandular tissues and result in autoimmune reactions that disrupt tear stability and lead to chronic dry eye disease. We here propose that unique features of the ocular surface immune system and the impact of systemic immune dysregulation in autoimmune diseases, can predispose to development of dry eye disease, and exacerbate severity of existing dry eye.


Assuntos
Doenças Autoimunes , Imunidade Inata , Ceratoconjuntivite Seca , Humanos , Ceratoconjuntivite Seca/imunologia , Doenças Autoimunes/imunologia , Túnica Conjuntiva/imunologia , Túnica Conjuntiva/patologia , Lágrimas/imunologia , Lágrimas/metabolismo
19.
Clin Immunol ; 147(2): 133-43, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23583916

RESUMO

Post-translational protein modifications can play a significant role in immune cell signaling. Recently, we showed that inhibition of transmethylation curtails experimental autoimmune encephalomyelitis, notably by reducing T cell receptor (TCR)-induced activation of CD4(+) T cells. Here, we demonstrate that transmethylation inhibition by a reversible S-adenosyl-l-homocysteine hydrolase inhibitor (DZ2002) led to immunosuppression by reducing TLR-, B cell receptor (BCR)- and TCR-induced activation of immune cells, most likely by blocking NF-κB activity. Moreover, prophylactic treatment with DZ2002 prevented lupus-like disease from developing in both BXSB and MRL-Fas(lpr) mouse models. DZ2002 treatment initiated during active disease significantly improved outcomes in both in vivo models, suggesting methylation inhibition as a novel approach for the treatment of autoimmune/inflammatory diseases.


Assuntos
Adenina/análogos & derivados , Butiratos/uso terapêutico , Imunossupressores/uso terapêutico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Receptores Toll-Like/imunologia , Adenina/uso terapêutico , Animais , Células Apresentadoras de Antígenos/imunologia , Autoanticorpos/sangue , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Imunoglobulina G/sangue , Rim/efeitos dos fármacos , Rim/patologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/imunologia , Processamento de Proteína Pós-Traducional , Transdução de Sinais
20.
Clin Immunol ; 143(1): 8-21, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22364920

RESUMO

The activation of immune cells is mediated by a network of signaling proteins that can undergo post-translational modifications critical for their activity. Methylation of nucleic acids or proteins can have major effects on gene expression as well as protein repertoire diversity and function. Emerging data indicate that indeed many immunologic functions, particularly those of T cells, including thymic education, differentiation and effector function are highly dependent on methylation events. The critical role of methylation in immunocyte biology is further documented by evidence that autoimmune phenomena may be curtailed by methylation inhibitors. Additionally, epigenetic alterations imprinted by methylation can also exert effects on normal and abnormal immune responses. Further work in defining methylation effects in the immune system is likely to lead to a more detailed understanding of the immune system and may point to the development of novel therapeutic approaches.


Assuntos
Autoimunidade/imunologia , Sistema Imunitário/imunologia , Imunidade/imunologia , Linfócitos T/imunologia , Animais , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/metabolismo , Metilação , Modelos Imunológicos , Transdução de Sinais/imunologia , Linfócitos T/citologia , Linfócitos T/metabolismo , Timócitos/citologia , Timócitos/imunologia , Timócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA