Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int Immunol ; 35(9): 403-421, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37227084

RESUMO

The interleukin-6 (IL-6) amplifier, which describes the simultaneous activation of signal transducer and activator of transcription 3 (STAT3) and NF-κb nuclear factor kappa B (NF-κB), in synovial fibroblasts causes the infiltration of immune cells into the joints of F759 mice. The result is a disease that resembles human rheumatoid arthritis. However, the kinetics and regulatory mechanisms of how augmented transcriptional activation by STAT3 and NF-κB leads to F759 arthritis is unknown. We here show that the STAT3-NF-κB complex is present in the cytoplasm and nucleus and accumulates around NF-κB binding sites of the IL-6 promoter region and established a computer model that shows IL-6 and IL-17 (interleukin 17) signaling promotes the formation of the STAT3-NF-κB complex followed by its binding on promoter regions of NF-κB target genes to accelerate inflammatory responses, including the production of IL-6, epiregulin, and C-C motif chemokine ligand 2 (CCL2), phenotypes consistent with in vitro experiments. The binding also promoted cell growth in the synovium and the recruitment of T helper 17 (Th17) cells and macrophages in the joints. Anti-IL-6 blocking antibody treatment inhibited inflammatory responses even at the late phase, but anti-IL-17 and anti-TNFα antibodies did not. However, anti-IL-17 antibody at the early phase showed inhibitory effects, suggesting that the IL-6 amplifier is dependent on IL-6 and IL-17 stimulation at the early phase, but only on IL-6 at the late phase. These findings demonstrate the molecular mechanism of F759 arthritis can be recapitulated in silico and identify a possible therapeutic strategy for IL-6 amplifier-dependent chronic inflammatory diseases.


Assuntos
Artrite Reumatoide , Interleucina-6 , Humanos , Animais , Camundongos , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Membrana Sinovial/metabolismo , Simulação por Computador , Fibroblastos/metabolismo
2.
Int Immunol ; 35(7): 313-326, 2023 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-36933193

RESUMO

Using a zoobiquity concept, we directly connect animal phenotypes to a human disease mechanism: the reduction of local plasminogen levels caused by matrix metalloproteinase-9 (MMP9) activity is associated with the development of inflammation in the intestines of dogs and patients with inflammatory bowel disease. We first investigated inflammatory colorectal polyps (ICRPs), which are a canine gastrointestinal disease characterized by the presence of idiopathic chronic inflammation, in Miniature Dachshund (MD) and found 31 missense disease-associated SNPs by whole-exome sequencing. We sequenced them in 10 other dog breeds and found five, PLG, TCOF1, TG, COL9A2 and COL4A4, only in MD. We then investigated two rare and breed-specific missense SNPs (T/T SNPs), PLG: c.477G > T and c.478A>T, and found that ICRPs with the T/T SNP risk alleles showed less intact plasminogen and plasmin activity in the lesions compared to ICRPs without the risk alleles but no differences in serum. Moreover, we show that MMP9, which is an NF-κB target, caused the plasminogen reduction and that intestinal epithelial cells expressing plasminogen molecules were co-localized with epithelial cells expressing MMP9 in normal colons with the risk alleles. Importantly, MMP9 expression in patients with ulcerous colitis or Crohn's disease also co-localized with epithelial cells showing enhanced NF-κB activation and less plasminogen expression. Overall, our zoobiquity experiments showed that MMP9 induces the plasminogen reduction in the intestine, contributing to the development of local inflammation and suggesting the local MMP9-plasminogen axis is a therapeutic target in both dogs and patients. Therefore, zoobiquity-type experiments could bring new perspectives for biomarkers and therapeutic targets.


Assuntos
Doenças Inflamatórias Intestinais , Metaloproteinase 9 da Matriz , Humanos , Cães , Animais , Plasminogênio , NF-kappa B , Inflamação , Serina Proteases
3.
Int Immunol ; 32(5): 335-346, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31930291

RESUMO

Chronic allograft rejection is the most common cause of long-term allograft failure. One reason is that current diagnostics and therapeutics for chronic allograft rejection are very limited. We here show that enhanced NFκB signaling in kidney grafts contributes to chronic active antibody-mediated rejection (CAAMR), which is a major pathology of chronic kidney allograft rejections. Moreover, we found that urinary orosomucoid 1 (ORM1) is a candidate marker molecule and therapeutic target for CAAMR. Indeed, urinary ORM1 concentration was significantly higher in kidney transplant recipients pathologically diagnosed with CAAMR than in kidney transplant recipients with normal histology, calcineurin inhibitor toxicity, or interstitial fibrosis and tubular atrophy. Additionally, we found that kidney biopsy samples with CAAMR expressed more ORM1 and had higher NFκB and STAT3 activation in tubular cells than samples from non-CAAMR samples. Consistently, ORM1 production was induced after cytokine-mediated NFκB and STAT3 activation in primary kidney tubular cells. The loss- and gain-of-function of ORM1 suppressed and promoted NFκB activation, respectively. Finally, ORM1-enhanced NFκB-mediated inflammation development in vivo. These results suggest that an enhanced NFκB-dependent pathway following NFκB and STAT3 activation in the grafts is involved in the development of chronic allograft rejection after kidney transplantation and that ORM1 is a non-invasive candidate biomarker and possible therapeutic target for chronic kidney allograft rejection.


Assuntos
Rejeição de Enxerto/imunologia , Nefropatias/imunologia , Transplante de Rim/efeitos adversos , Orosomucoide/metabolismo , Animais , Biomarcadores/análise , Linhagem Celular , Doença Crônica , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/terapia , Humanos , Nefropatias/diagnóstico , Nefropatias/terapia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Orosomucoide/análise , Orosomucoide/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia , Transplante Homólogo/efeitos adversos
4.
J Immunol ; 201(8): 2264-2272, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30209188

RESUMO

Bmi1 is a polycomb group protein and regulator that stabilizes the ubiquitination complex PRC1 in the nucleus with no evidently direct link to the NF-κB pathway. In this study, we report a novel function of Bmi1: its regulation of IκBα ubiquitination in the cytoplasm. A deficiency of Bmi1 inhibited NF-κB-mediated gene expression in vitro and a NF-κB-mediated mouse model of arthritis in vivo. Mechanistic analysis showed that Bmi1 associated with the SCF ubiquitination complex via its N terminus and with phosphorylation by an IKKα/ß-dependent pathway, leading to the ubiquitination of IκBα. These effects on NF-κB-related inflammation suggest Bmi1 in the SCF complex is a potential therapeutic target for various diseases and disorders, including autoimmune diseases.


Assuntos
Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Citoplasma/metabolismo , Células Endoteliais/fisiologia , Complexos Multiproteicos/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/genética , NF-kappa B/metabolismo , Complexo Repressor Polycomb 1/genética , Ligação Proteica , Estabilidade Proteica , Proteólise , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/genética , Proteínas Ligases SKP Culina F-Box/genética , Ativação Transcricional , Ubiquitinação
5.
Int Immunol ; 29(12): 581-591, 2017 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-29309623

RESUMO

RNA-binding motif 10 (Rbm10) is an RNA-binding protein that regulates alternative splicing, but its role in inflammation is not well defined. Here, we show that Rbm10 controls appropriate splicing of DNA (cytosine-5)-methyltransferase 3b (Dnmt3b), a DNA methyltransferase, to regulate the activity of NF-κB-responsive promoters and consequently inflammation development. Rbm10 deficiency suppressed NF-κB-mediated responses in vivo and in vitro. Mechanistic analysis showed that Rbm10 deficiency decreased promoter recruitment of NF-κB, with increased DNA methylation of the promoter regions in NF-κB-responsive genes. Consistently, Rbm10 deficiency increased the expression level of Dnmt3b2, which has enzyme activity, while it decreased the splicing isoform Dnmt3b3, which does not. These two isoforms associated with NF-κB efficiently, and overexpression of enzymatically active Dnmt3b2 suppressed the expression of NF-κB targets, indicating that Rbm10-mediated Dnmt3b2 regulation is important for the induction of NF-κB-mediated transcription. Therefore, Rbm10-dependent Dnmt3b regulation is a possible therapeutic target for various inflammatory diseases.


Assuntos
Artrite/imunologia , DNA (Citosina-5-)-Metiltransferases/genética , Inflamação/imunologia , Isoformas de Proteínas/genética , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo/genética , Animais , Artrite/genética , Células Cultivadas , Modelos Animais de Doenças , Humanos , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Ativação Transcricional , DNA Metiltransferase 3B
6.
J Immunol ; 197(8): 3111-3119, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27630163

RESUMO

The breakpoint cluster region (BCR) is known as a kinase and cause of leukemia upon fusing to Abl kinase. In this study, we demonstrate that BCR associated with the α subunit of casein kinase II (CK2α), rather than BCR itself, is required for inflammation development. We found that BCR knockdown inhibited NF-κB activation in vitro and in vivo. Computer simulation, however, suggested that the putative BCR kinase domain has an unstable structure with minimal enzymatic activity. Liquid chromatography-tandem mass spectrometry analysis showed that CK2α associated with BCR. We found the BCR functions are mediated by CK2α. Indeed, CK2α associated with adaptor molecules of TNF-αR and phosphorylated BCR at Y177 to establish a p65 binding site after TNF-α stimulation. Notably, p65 S529 phosphorylation by CK2α creates a p300 binding site and increased p65-mediated transcription followed by inflammation development in vivo. These results suggest that BCR-mediated inflammation is dependent on CK2α, and the BCR-CK2α complex could be a novel therapeutic target for various inflammatory diseases.


Assuntos
Artrite/genética , Caseína Quinase II/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Cromossomo Filadélfia , Proteínas Proto-Oncogênicas c-bcr/metabolismo , Animais , Artrite Experimental/genética , Linhagem Celular , Cromatografia Líquida , Proteínas de Fusão bcr-abl/genética , Genes abl/genética , Humanos , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcr/genética , RNA Interferente Pequeno/genética , Espectrometria de Massas em Tandem , Fator de Necrose Tumoral alfa/metabolismo
7.
Int Immunol ; 28(3): 117-26, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26489882

RESUMO

KDEL receptor 1 (KDELR1) regulates integrated stress responses (ISR) to promote naive T-cell survival in vivo. In a mouse line having nonfunctional KDELR1, T-Red (naive T-cell reduced) mice, polyclonal naive T cells show excessive ISR and eventually undergo apoptosis. However, breeding T-Red mice with TCR-transgenic mice bearing relatively high TCR affinity rescued the T-Red phenotype, implying a link between ISR-induced apoptosis and TCR-mediated signaling. Here, we showed that strong TCR stimulation reduces ISR in naive T cells. In mice lacking functional KDELR1, surviving naive T cells expressed significantly higher levels of CD5, a surrogate marker of TCR self-reactivity. In addition, higher TCR affinity/avidity was confirmed using a tetramer dissociation assay on the surviving naive T cells, suggesting that among the naive T-cell repertoire, those that receive relatively stronger TCR-mediated signals via self-antigens survive enhanced ISR. Consistent with this observation, weak TCR stimulation with altered peptide ligands decreased the survival and proliferation of naive T cells, whereas stimulation with ligands having higher affinity had no such effect. These results suggest a novel role of TCR-mediated signals in the attenuation of ISR in vivo.


Assuntos
Estresse do Retículo Endoplasmático/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Peptídeos/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/fisiologia , Animais , Apoptose/genética , Autoantígenos/imunologia , Antígenos CD5/metabolismo , Sobrevivência Celular/genética , Células Cultivadas , Estresse do Retículo Endoplasmático/genética , Homeostase/genética , Humanos , Memória Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Peptídeos/genética , Transdução de Sinais/genética
8.
J Immunol ; 194(3): 1039-46, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25556244

RESUMO

In this study, we investigated the relationship between several growth factors and inflammation development. Serum concentrations of epiregulin, amphiregulin, betacellulin, TGF-α, fibroblast growth factor 2, placental growth factor (PLGF), and tenascin C were increased in rheumatoid arthritis patients. Furthermore, local blockades of these growth factors suppressed the development of cytokine-induced arthritis in mice by inhibiting chemokine and IL-6 expressions. We found that epiregulin expression was early and followed by the induction of other growth factors at different sites of the joints. The same growth factors then regulated the expression of epiregulin at later time points of the arthritis. These growth factors were increased in patients suffering from multiple sclerosis (MS) and also played a role in the development of an MS model, experimental autoimmune encephalomyelitis. The results suggest that the temporal expression of growth factors is involved in the inflammation development seen in several diseases, including rheumatoid arthritis and MS. Therefore, various growth factor pathways might be good therapeutic targets for various inflammatory diseases.


Assuntos
Epirregulina/metabolismo , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Citocinas/sangue , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Humanos , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Camundongos , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
9.
Proc Natl Acad Sci U S A ; 111(32): 11786-91, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25074919

RESUMO

The humoral immune response, also called the antibody-mediated immune response, is one of the main adaptive immune systems. The essential micronutrient zinc (Zn) is known to modulate adaptive immune responses, and dysregulated Zn homeostasis leads to immunodeficiency. However, the molecular mechanisms underlying this Zn-mediated modulation are largely unknown. Here, we show that the Zn transporter SLC39A10/ZIP10 plays an important role in B-cell antigen receptor (BCR) signal transduction. Zip10-deficiency in mature B cells attenuated both T-cell-dependent and -independent immune responses in vivo. The Zip10-deficient mature B cells proliferated poorly in response to BCR cross-linking, as a result of dysregulated BCR signaling. The perturbed signaling was found to be triggered by a reduction in CD45R phosphatase activity and consequent hyperactivation of LYN, an essential protein kinase in BCR signaling. Our data suggest that ZIP10 functions as a positive regulator of CD45R to modulate the BCR signal strength, thereby setting a threshold for BCR signaling in humoral immune responses.


Assuntos
Proteínas de Transporte de Cátions/imunologia , Imunidade Humoral , Receptores de Antígenos de Linfócitos B/metabolismo , Zinco/metabolismo , Imunidade Adaptativa , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Diferenciação Celular/imunologia , Senescência Celular/imunologia , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/imunologia , Linfócitos T/imunologia
11.
Crit Rev Immunol ; 35(5): 365-78, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26853849

RESUMO

Inflammation is a fundamental response induced by the immune system to protect the body against pathogens, tissue damage, and stress. At the same time, recent studies have suggested that chronically induced inflammation is involved in various human diseases and disorders. Thus, understanding the molecular mechanisms of chronic inflammation could provide therapeutic value. Many mediators such as cytokines or chemokines regulate inflammatory responses. Among them, interleukin(IL)-6 is a prominent cytokine that induces and maintains inflammatory reactions. It is expressed by activated CD4+ T cells and also non-immune cells such as fibroblasts and epithelial cells. We discovered an inflammation-induction machinery, the inflammation amplifier, which is activated by the simultaneous stimulation of nuclear factor-kappa B (NF-κB) and signal transducers and activator of transcription 3 (STAT3) via various cytokines like IL-17 and IL-6 in non-immune cells. Activation of the inflammation amplifier induces a synergistic increase of IL-6, inflammatory chemokines, and growth factors. Using genome-wide screening, we identified several growth factors as mediators of the inflammation amplifier. In this review, we highlight the role of growth factors in the inflammation mechanism with special attention on the inflammation amplifier.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doenças do Sistema Imunitário/imunologia , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/imunologia , Animais , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Interleucina-17/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
12.
Nihon Rinsho ; 73(4): 693-700, 2015 Apr.
Artigo em Japonês | MEDLINE | ID: mdl-25936164

RESUMO

Inflammation is observed in many diseases and disorders. We discovered a key machinery of inflammation, the inflammation amplifier, which is induced by the simultaneous activation of NFκB and STAT3 followed by the hyper-activation of NFκB in non-immune cells, including endothelial cells and fibroblasts. Since that discovery, we found the Gateway Reflex, which describes regional neural activations that enhance the inflammation amplifier to create a gateway for immune cells to bypass the blood-brain barrier. In addition, we have identified over 1,000 positive regulators and over 500 targets of the inflammation amplifier, which include a significant numbers of human disease-associated genes. In parallel, we performed a comprehensive analysis of human disease samples and found that the inflammation amplifier was activated during the development of chronic inflammation. Thus, we concluded that the inflammation amplifier is associated with various human diseases and disorders, including autoimmune diseases, metabolic syndromes, neurodegenerative diseases, and other inflammatory diseases. We are now attempting drug discovery for inflammatory diseases and disorders based on the inflammation amplifier and Gateway Reflex. In this review, we discuss the Gateway Reflex as an example for the neuro-immune interaction in vivo.


Assuntos
Sistema Nervoso Central/imunologia , Inflamação/imunologia , Animais , Doenças Autoimunes/imunologia , Sistema Nervoso Central/metabolismo , Ciclinas/imunologia , Humanos , Reflexo , Transdução de Sinais
14.
Mediators Inflamm ; 2013: 898165, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23990699

RESUMO

The central nervous system (CNS) is an immune-privileged environment protected by the blood-brain barrier (BBB), which consists of specific endothelial cells that are brought together by tight junctions and tight liner sheets formed by pericytes and astrocytic end-feet. Despite the BBB, various immune and tumor cells can infiltrate the CNS parenchyma, as seen in several autoimmune diseases like multiple sclerosis (MS), cancer metastasis, and virus infections. Aside from a mechanical disruption of the BBB like trauma, how and where these cells enter and accumulate in the CNS from the blood is a matter of debate. Recently, using experimental autoimmune encephalomyelitis (EAE), an animal model of MS, we found a "gateway" at the fifth lumber cord where pathogenic autoreactive CD4+ T cells can cross the BBB. Interestingly, this gateway is regulated by regional neural stimulations that can be mechanistically explained by the gate theory. In this review, we also discuss this theory and its potential for treating human diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Sistema Nervoso Central/imunologia , Neurônios/metabolismo , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/fisiologia , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Células Endoteliais/citologia , Humanos , Inflamação/patologia , Camundongos , Modelos Biológicos , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Medula Espinal/patologia
15.
Int Immunol ; 22(5): 375-86, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20215335

RESUMO

Zinc (Zn) is an essential trace metal required by many enzymes and transcription factors for their activity or the maintenance of their structure. Zn has a variety of effects in the immune responses and inflammation, although it has not been well known how Zn affects these reactions on the molecular basis. We here showed that Zn suppresses T(h)17-mediated autoimmune diseases at lest in part by inhibiting the development of T(h)17 cells via attenuating STAT3 activation. In mice injected with type II collagen to induce arthritis, Zn treatment inhibited T(h)17 cell development. IL-6-mediated activation of STAT3 and in vitro T(h)17 cell development were all suppressed by Zn. Importantly, Zn binding changed the alpha-helical secondary structure of STAT3, disrupting the association of STAT3 with JAK2 kinase and with a phospho-peptide that included a STAT3-binding motif from the IL-6 signal transducer gp130. Thus, we conclude that Zn suppresses STAT3 activation, which is a critical step for T(h)17 development.


Assuntos
Artrite Experimental/tratamento farmacológico , Interleucina-17/imunologia , Fator de Transcrição STAT3/antagonistas & inibidores , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Zinco/farmacologia , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/metabolismo , Relação Estrutura-Atividade , Células Th17/citologia , Células Th17/metabolismo
16.
J Immunol ; 182(12): 7527-38, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494276

RESUMO

The tripartite motif (TRIM) family member, TRIM21, is an E3 ubiquitin ligase for IFN regulatory factor (IRF)3 and IRF8 that functions in both innate and acquired immunity. It is also an autoantigen known as Ro52/SS-A. The function of TRIM21 in vivo, however, has remained elusive. We generated Trim21(-/-) mice with the Trim21 gene replaced by an enhanced GFP (EGFP) reporter. EGFP expression analyses showed that Trim21 was widely expressed in many tissues, with the highest levels in immune cells. Studies of Trim21(-/-) embryonic fibroblasts demonstrated that TLR-mediated induction of proinflammatory cytokines, including IL-1beta, IL-6, TNF-alpha, and CXCL10, was consistently up-regulated relative to wild-type cells. Reporter analyses demonstrated that TLR-mediated NF-kappaB activation was higher in Trim21(-/-) cells than in wild-type cells, most likely accounting for their enhanced cytokine expression. In contrast, functional analyses of immune cells from Trim21(-/-) mice revealed no abnormalities in their composition or function, even though ubiquitylation of IRF3 and IRF8 was impaired. These results suggested possible redundancies in activities mediated by TRIM21. In keeping with this concept, we found that a number of TRIM family members were up-regulated in Trim21(-/-) cells. Taken together, these findings demonstrate that TRIM21 plays a previously unrecognized role in the negative regulation of NF-kappaB-dependent proinflammatory cytokine responses, and suggest that multiple TRIM proteins contribute to the maintenance of functional equilibrium in inflammatory responses, in part through functional redundancy.


Assuntos
Citocinas/imunologia , Citocinas/metabolismo , NF-kappa B/metabolismo , Ribonucleoproteínas/metabolismo , Animais , Medula Óssea/imunologia , Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fibroblastos , Regulação da Expressão Gênica , Genes Reporter/genética , Ativação Linfocitária/imunologia , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Especificidade de Órgãos , Reprodução , Ribonucleoproteínas/deficiência , Ribonucleoproteínas/genética , Transdução de Sinais/imunologia , Taxa de Sobrevida , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Ubiquitinação
17.
J Exp Med ; 196(7): 979-90, 2002 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-12370259

RESUMO

We generated a mouse line in which the src homology 2 domain-bearing protein tyrosine phosphatase (SHP)-2 binding site of gp130, tyrosine 759, was mutated to phenylalanine (gp130(F759/F759)). The gp130(F759/F759) mice developed rheumatoid arthritis (RA)-like joint disease. The disease was accompanied by autoantibody production and accumulated memory/activated T cells and myeloid cells. Before the disease onset, the T cells were hyperresponsive and thymic selection and peripheral clonal deletion were impaired. The inhibitory effect of IL-6 on Fas ligand expression during activation-induced cell death (AICD) was augmented in gp130(F759/F759) T cells in a manner dependent on the tyrosine residues of gp130 required for signal transducer and activator of transcription 3 activation. Finally, we showed that disease development was dependent on lymphocytes. These results provide evidence that a point mutation of a cytokine receptor has the potential to induce autoimmune disease.


Assuntos
Artrite Experimental/imunologia , Receptores de Interleucina-6/genética , Linfócitos T/imunologia , Tirosina , Substituição de Aminoácidos , Animais , Formação de Anticorpos , Apoptose , Artrite Experimental/genética , Artrite Experimental/patologia , Sequência de Bases , Sítios de Ligação , Osso e Ossos/patologia , Linfócitos T CD4-Positivos/imunologia , Primers do DNA , Enterotoxinas/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação Puntual , Reação em Cadeia da Polimerase , Subunidades Proteicas , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Linfócitos T/fisiologia
18.
Blood ; 112(10): 4028-38, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18799728

RESUMO

PU.1, IKAROS, E2A, EBF, and PAX5 comprise a transcriptional network that orchestrates B-cell lineage specification, commitment, and differentiation. Here we identify interferon regulatory factor 8 (IRF8) as another component of this complex, and show that it also modulates lineage choice by hematopoietic stem cells (HSCs). IRF8 binds directly to an IRF8/Ets consensus sequence located in promoter regions of Sfpi1 and Ebf1, which encode PU.1 and EBF, respectively, and is associated with transcriptional repression of Sfpi1 and transcriptional activation of Ebf1. Bone marrows of IRF8 knockout mice (IRF8(-/-)) had significantly reduced numbers of pre-pro-B cells and increased numbers of myeloid cells. Although HSCs of IRF8(-/-) mice failed to differentiate to B220(+) B-lineage cells in vitro, the defect could be rescued by transfecting HSCs with wild-type but not with a signaling-deficient IRF8 mutant. In contrast, overexpression of IRF8 in HSC-differentiated progenitor cells resulted in growth inhibition and apoptosis. We also found that IRF8 was expressed at higher levels in pre-pro-B cells than more mature B cells in wild-type mice. Together, these results indicate that IRF8 modulates lineage choice by HSCs and is part of the transcriptional network governing B-cell lineage specification, commitment, and differentiation.


Assuntos
Linfócitos B/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Fatores Reguladores de Interferon/metabolismo , Elementos de Resposta/fisiologia , Transdução de Sinais/fisiologia , Animais , Linfócitos B/citologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células-Tronco Hematopoéticas/química , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Fatores Reguladores de Interferon/genética , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Knockout , Fator de Transcrição PAX5/genética , Fator de Transcrição PAX5/metabolismo , Ligação Proteica/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/genética , Transativadores/metabolismo
19.
Int Immunol ; 21(1): 73-80, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19050105

RESUMO

Infection with pathogens containing superantigens (Sags) can result in massive excessive CD4+ T cell activation and death in such conditions as toxic shock, food poisoning and autoimmune diseases. We here showed how enhancement of IL-6 signaling suppresses Sag-mediated activated CD4+ T cell death. Sag-induced CD4+ T cell death increased in IL-6 knockout (KO) mice, whereas it decreased in mice characterized by enhanced IL-6-gp130-STAT3 signaling. The serum concentration of IFN-gamma was inversely correlated with the magnitude of IL-6 signaling, and IFN-gamma deficiency inhibited Sag-induced activated CD4+ T cell death, suggesting that IL-6 suppresses CD4+ T cell death via IFN-gamma expression. Interestingly, depletion of activated CD8+ T cells inhibited Sag-mediated increases in IFN-gamma expression in IL-6 KO mice as well as the augmented CD4+ T cell death. The results demonstrate that IL-6-gp130-STAT3 signaling in activated CD8+ T cells contributes to Sag-induced CD4+ T cell death via IFN-gamma expression, highlighting this signaling axis in CD8+ T cells as a potential therapeutic target for Sag-related syndromes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/metabolismo , Interleucina-6/metabolismo , Superantígenos/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Interferon gama/sangue , Interleucina-6/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Superantígenos/metabolismo
20.
J Invest Dermatol ; 139(2): 333-341, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30273597

RESUMO

Keloids mark a chronic inflammatory disease characterized by a fibroproliferative disorder of the skin. A genome-wide association study showed that single-nucleotide polymorphism rs8032158 in the neural precursor cell-expressed NEDD4 gene, which has six protein-coding transcript variants (TVs), is genetically linked to keloids. Here, we show that the high frequency of risk allele C in rs8032158 in keloid patients is associated with a selectively higher expression of TV3 of NEDD4 to activate the NF-κB pathway. Comparisons of keloid scars with normal skin samples that do not have the single-nucleotide polymorphism allele and were derived from different anatomical sites showed stronger expressions of NEDD4 TV3 and activated forms of NF-κB and STAT3 in keloid scars. Forced expression or selective knockdown of NEDD4 TV3 increased or decreased NF-κB activation in vitro. Furthermore, NEDD4 knockdown suppressed NF-κB-dependent inflammation development in vivo. Mechanistic analysis showed that NEDD4 TV3 is involved in NF-κB activation through its association with the adaptor protein RIP. These results suggest that NEDD4 TV3 is a potential diagnostic marker and therapeutic target for chronic skin diseases, including keloid.


Assuntos
Inflamação/patologia , Queloide/patologia , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Pele/patologia , Adulto , Idoso , Alelos , Biomarcadores/metabolismo , Linhagem Celular , Criança , Feminino , Fibroblastos , Técnicas de Silenciamento de Genes , Humanos , Lactente , Inflamação/genética , Queloide/genética , Queratinócitos , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Polimorfismo de Nucleotídeo Único , Cultura Primária de Células , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Pele/citologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA