Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell ; 185(18): 3375-3389.e21, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35998627

RESUMO

Systemic lupus erythematosus (SLE) is a complex autoimmune disease involving multiple immune cells. To elucidate SLE pathogenesis, it is essential to understand the dysregulated gene expression pattern linked to various clinical statuses with a high cellular resolution. Here, we conducted a large-scale transcriptome study with 6,386 RNA sequencing data covering 27 immune cell types from 136 SLE and 89 healthy donors. We profiled two distinct cell-type-specific transcriptomic signatures: disease-state and disease-activity signatures, reflecting disease establishment and exacerbation, respectively. We then identified candidate biological processes unique to each signature. This study suggested the clinical value of disease-activity signatures, which were associated with organ involvement and therapeutic responses. However, disease-activity signatures were less enriched around SLE risk variants than disease-state signatures, suggesting that current genetic studies may not well capture clinically vital biology. Together, we identified comprehensive gene signatures of SLE, which will provide essential foundations for future genomic and genetic studies.


Assuntos
Lúpus Eritematoso Sistêmico , Transcriptoma , Humanos , Lúpus Eritematoso Sistêmico/genética , Análise de Sequência de RNA
2.
Cell ; 184(11): 3006-3021.e17, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33930287

RESUMO

Genetic studies have revealed many variant loci that are associated with immune-mediated diseases. To elucidate the disease pathogenesis, it is essential to understand the function of these variants, especially under disease-associated conditions. Here, we performed a large-scale immune cell gene-expression analysis, together with whole-genome sequence analysis. Our dataset consists of 28 distinct immune cell subsets from 337 patients diagnosed with 10 categories of immune-mediated diseases and 79 healthy volunteers. Our dataset captured distinctive gene-expression profiles across immune cell types and diseases. Expression quantitative trait loci (eQTL) analysis revealed dynamic variations of eQTL effects in the context of immunological conditions, as well as cell types. These cell-type-specific and context-dependent eQTLs showed significant enrichment in immune disease-associated genetic variants, and they implicated the disease-relevant cell types, genes, and environment. This atlas deepens our understanding of the immunogenetic functions of disease-associated variants under in vivo disease conditions.


Assuntos
Regulação da Expressão Gênica/genética , Expressão Gênica/imunologia , Doenças do Sistema Imunitário/genética , Adulto , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla/métodos , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/metabolismo , Doenças do Sistema Imunitário/metabolismo , Doenças do Sistema Imunitário/fisiopatologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Locos de Características Quantitativas/genética , Locos de Características Quantitativas/imunologia , Transcriptoma/genética , Sequenciamento Completo do Genoma/métodos
3.
Ann Rheum Dis ; 82(6): 809-819, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36918189

RESUMO

OBJECTIVES: Little is known about the immunology underlying variable treatment response in rheumatoid arthritis (RA). We performed large-scale transcriptome analyses of peripheral blood immune cell subsets to identify immune cells that predict treatment resistance. METHODS: We isolated 18 peripheral blood immune cell subsets of 55 patients with RA requiring addition of new treatment and 39 healthy controls, and performed RNA sequencing. Transcriptome changes in RA and treatment effects were systematically characterised. Association between immune cell gene modules and treatment resistance was evaluated. We validated predictive value of identified parameters for treatment resistance using quantitative PCR (qPCR) and mass cytometric analysis cohorts. We also characterised the identified population by synovial single cell RNA-sequencing analysis. RESULTS: Immune cells of patients with RA were characterised by enhanced interferon and IL6-JAK-STAT3 signalling that demonstrate partial normalisation after treatment. A gene expression module of plasmacytoid dendritic cells (pDC) reflecting the expansion of dendritic cell precursors (pre-DC) exhibited strongest association with treatment resistance. Type I interferon signalling was negatively correlated to pre-DC gene expression. qPCR and mass cytometric analysis in independent cohorts validated that the pre-DC associated gene expression and the proportion of pre-DC were significantly higher before treatment in treatment-resistant patients. A cluster of synovial DCs showed both features of pre-DC and pro-inflammatory conventional DC2s. CONCLUSIONS: An increase in pre-DC in peripheral blood predicted RA treatment resistance. Pre-DC could have pathophysiological relevance to RA treatment response.


Assuntos
Artrite Reumatoide , Humanos , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/genética , Transcriptoma , Perfilação da Expressão Gênica , Células Dendríticas
4.
Rheumatology (Oxford) ; 62(2): 905-913, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35689621

RESUMO

OBJECTIVES: To investigate metabolite alterations in the plasma of SLE patients to identify novel biomarkers and provide insight into SLE pathogenesis. METHODS: Patients with SLE (n = 41, discovery cohort and n = 37, replication cohort), healthy controls (n = 30 and n = 29) and patients with RA (n = 19, disease control) were recruited. Metabolic profiles of the plasma samples were analysed using liquid chromatography-time-of-flight mass spectrometry and capillary electrophoresis-time-of-flight mass spectrometry. Transcriptome data was analysed using RNA-sequencing for 18 immune cell subsets. The importance of histidine (His) in plasmablast differentiation was investigated by using mouse splenic B cells. RESULTS: We demonstrate that a specific amino acid combination including His can effectively distinguish between SLE patients and healthy controls. Random forest and partial least squares-discriminant analysis identified His as an effective classifier for SLE patients. A decrease in His plasma levels correlated with damage accrual independent of prednisolone dosage and type I IFN signature. The oxidative phosphorylation signature in plasmablasts negatively correlated with His levels. We also showed that plasmablast differentiation induced by innate immune signals was dependent on His. CONCLUSIONS: Plasma His levels are a potential biomarker for SLE patients and are associated with damage accrual. Our data suggest the importance of His as a pathogenic metabolite in SLE pathogenesis.


Assuntos
Histidina , Lúpus Eritematoso Sistêmico , Animais , Camundongos , Transcriptoma , Metabolômica/métodos , Biomarcadores , Lúpus Eritematoso Sistêmico/genética
5.
Ann Rheum Dis ; 81(6): 845-853, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35236659

RESUMO

OBJECTIVE: Systemic lupus erythematosus (SLE) is the prototypical systemic autoimmune disease. While the long-term prognosis has greatly improved, better long-term survival is still necessary. The type I interferon (IFN) signature, a prominent feature of SLE, is not an ideal therapeutic target or outcome predictor. To explore immunological pathways in SLE more precisely, we performed transcriptomic, epigenomic and genomic analyses using 19 immune cell subsets from peripheral blood. METHODS: We sorted 19 immune cell subsets and identified the mRNA expression profiles and genetic polymorphisms in 107 patients with SLE and 92 healthy controls. Combined differentially expressed genes and expression quantitative trait loci analysis was conducted to find key driver genes in SLE pathogenesis. RESULTS: We found transcriptomic, epigenetic and genetic importance of oxidative phosphorylation (OXPHOS)/mitochondrial dysfunction in SLE memory B cells. Particularly, we identified an OXPHOS-regulating gene, PRDX6 (peroxiredoxin 6), as a key driver in SLE B cells. Prdx6-deficient B cells showed upregulated mitochondrial respiration as well as antibody production. We revealed OXPHOS signature was associated with type I IFN signalling-related genes (ISRGs) signature in SLE memory B cells. Furthermore, the gene sets related to innate immune signalling among ISRGs presented correlation with OXPHOS and these two signatures showed associations with SLE organ damage as well as specific clinical phenotypes. CONCLUSION: This work elucidated the potential prognostic marker for SLE. Since OXPHOS consists of the electron transport chain, a functional unit in mitochondria, these findings suggest the importance of mitochondrial dysfunction as a key immunological pathway involved in SLE.


Assuntos
Interferon Tipo I , Lúpus Eritematoso Sistêmico , Linfócitos B/metabolismo , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Fosforilação Oxidativa , Transcriptoma
6.
J Autoimmun ; 133: 102907, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36126366

RESUMO

OBJECTIVE: Human Leukocyte Antigen (HLA) alleles regulate susceptibility to rheumatoid arthritis (RA) and immune-mediated diseases. This study aims to elucidate the impact of HLA alleles to T cell subsets. METHODS: We performed genome-wide and HLA allele association analysis for T cell receptor (TCR) beta chain repertoire in 13 purified T cell subsets from the ImmuNexUT database, consisting of 407 donors with ten immune-mediated diseases and healthy controls. RESULTS: HLA class II alleles were associated with TRBV gene usage and the public clones of CD4 T cells, while HLA class I alleles were associated with CD8 T cells. RA-risk and immune-mediated diseases-risk HLA alleles were associated with TRBV gene usage of naive and effector CD4 T cell subsets and public clones accumulating in Th17. Clonal diversity was independent of HLA alleles and was correlated with transcriptome changes that reflect TCR signaling. CONCLUSION: This study revealed in vivo evidence that both HLA alleles and environmental factors shape naive and effector TCR repertoires in RA and immune-mediated diseases patients.


Assuntos
Artrite Reumatoide , Linfócitos T CD4-Positivos , Humanos , Artrite Reumatoide/genética , Receptores de Antígenos de Linfócitos T/genética
7.
Rheumatology (Oxford) ; 61(10): 4163-4174, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-35040949

RESUMO

OBJECTIVES: We evaluated flow-cytometric and transcriptome features of peripheral blood immune cells from early-phase (disease duration <5 years) SSc in comparison with late-phase SSc. METHODS: Fifty Japanese patients with SSc (12 early SSc cases and 38 late SSc cases) and 50 age- and sex-matched healthy controls were enrolled. A comparison of flow-cytometric subset proportions and RNA-sequencing of 24 peripheral blood immune cell subsets was performed. We evaluated differentially expressed genes (DEGs), characterized the co-expressed gene modules, and estimated the composition of subpopulations by deconvolution based on single-cell RNA-sequencing data. As a disease control, idiopathic inflammatory myositis (IIM) patients were also evaluated. RESULTS: Analysing the data from early and late SSc, fraction II effector regulatory T cell (Fr. II eTreg) genes showed a remarkable differential gene expression, enriched for genes related to oxidative phosphorylation. Although the flow-cytometric proportion of Fr. II eTregs was not changed in early SSc, deconvolution indicated expansion of the activated subpopulation. Co-expressed gene modules of Fr. II eTregs demonstrated enrichment of the DEGs of early SSc and correlation with the proportion of the activated subpopulation. These results suggested that DEGs in Fr. II eTregs from patients with early SSc were closely associated with the increased proportion of the activated subpopulation. Similar dysregulation of Fr. II eTregs was also observed in data from patients with early IIM. CONCLUSIONS: RNA-seq of immune cells indicated the dysregulation of Fr. II eTregs in early SSc with increased proportion of the activated subpopulation.


Assuntos
Escleroderma Sistêmico , Linfócitos T Reguladores , Citometria de Fluxo , Humanos , RNA , Análise de Sequência de RNA
8.
Rheumatology (Oxford) ; 61(11): 4445-4454, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-35179548

RESUMO

OBJECTIVES: To assess the efficacy and safety of branched chain amino acids (BCAAs) in the treatment of PM/DM prior to official approval of their use in Japan. METHODS: Treatment naïve adults with PM/DM were enrolled in a randomized, double-blind trial to receive either TK-98 (drug name of BCAAs) or placebo in addition to conventional treatment. After 12 weeks, patients with an average manual muscle test (MMT) score <9.5 were enrolled in an open label extension study for a further 12 weeks. The primary endpoint was the change of the MMT score at 12 weeks. The secondary endpoints were the clinical response and the change of functional index (FI). RESULTS: Forty-seven patients were randomized either to the TK-98 (n = 24) or placebo (n = 23) group. The changes of MMT scores at 12 weeks were 0.70 (0.19) [mean (s.e.m.)] and 0.69 (0.18), respectively (P = 0.98). Thirteen patients from the TK-98 group and 12 from the placebo group were enrolled in the extension study. The MMT scores in both groups improved similarly. The increase of the FI scores of the shoulder flexion at 12 weeks was significantly greater in the TK-98 group [27.9 (5.67) vs 12.8 (5.67) for the right shoulder flexion, and 27.0 (5.44) vs 13.4 (5.95) for the left shoulder; P < 0.05]. Frequencies of adverse events up to 12 weeks were similar. CONCLUSION: BCAAs showed no effect on the improvement of the muscle strength evaluated by MMT and the clinical response. However, they were partly effective for improving dynamic repetitive muscle functions. TRIAL REGISTRATION: UMIN-CTR Clinical Trial, https://center6.umin.ac.jp/, UMIN000016233.


Assuntos
Dermatomiosite , Polimiosite , Adulto , Humanos , Aminoácidos de Cadeia Ramificada/uso terapêutico , Dermatomiosite/tratamento farmacológico , Método Duplo-Cego , Força Muscular , Polimiosite/tratamento farmacológico , Resultado do Tratamento
9.
Rheumatol Int ; 42(10): 1863-1872, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34156527

RESUMO

Protein-losing enteropathy (PLE) has been reported to be associated with various systemic autoimmune diseases. However, reports regarding PLE in ANCA-associated vasculitis (AAV) patients are limited. We herein aimed to describe the clinical characteristics of AAV with PLE. We conducted a retrospective chart review of patients who were diagnosed with AAV and who began treatment at the University of Tokyo Hospital between June 2003 and June 2020. Among 68 AAV patients, there were four patients (5.9%) with PLE, consisting of two patients with MPA, one patient with GPA, and one patient with EGPA. Clinical courses were described, and their data were compared with AAV patients without PLE. Demographic characteristics, disease activity, and the pattern of organ involvement were similar between patients with PLE and without PLE. Patients with PLE had hypocomplementemia more frequently than the patients without PLE (CH50 75.0% vs 1.8%, p < 0.001, C3 50.0% vs 1.8%, p = 0.01, C4 75.0% vs 3.5%, p = 0.001). Although hypoalbuminemia improved with immunosuppressive therapy for AAV, the improvement in hypoalbuminemia was slow in most cases. We also performed a systematic review on PLE associated with vasculitis. Thirteen reports were included, and Henoch-Schonlein Purpura patients with PLE also tended to have hypocomplementemia. In conclusion, PLE is a rare complication of AAV and complement system may associate with the mechanism of PLE.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos , Hipoalbuminemia , Enteropatias Perdedoras de Proteínas , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/complicações , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/diagnóstico , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/tratamento farmacológico , Anticorpos Anticitoplasma de Neutrófilos , Proteínas do Sistema Complemento , Humanos , Hipoalbuminemia/complicações , Enteropatias Perdedoras de Proteínas/diagnóstico , Enteropatias Perdedoras de Proteínas/etiologia , Enteropatias Perdedoras de Proteínas/terapia , Estudos Retrospectivos
10.
J Autoimmun ; 116: 102547, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33039247

RESUMO

OBJECTIVE: Immunological disturbances have been reported in systemic sclerosis (SSc). This study assessed the transcriptome disturbances in immune cell subsets in SSc and characterized a disease-related gene network module and immune cell cluster at single cell resolution. METHODS: Twenty-one Japanese SSc patients were enrolled and compared with 13 age- and sex-matched healthy controls (HC). Nineteen peripheral blood immune cell subsets were sorted by flow cytometry and bulk RNA-seq analysis was performed for each. Differential expression and pathway analyses were conducted. Iterative weighted gene correlation network analysis (iWGCNA) of each subset revealed clustered co-expressed gene network modules. Random forest analysis prioritized a disease-related gene module. Single cell RNA-seq analysis of 878 monocytes was integrated with bulk RNA-seq analysis and with a public database for single cell RNA-seq analysis of SSc patients. RESULTS: Inflammatory pathway genes were differentially expressed in widespread immune cell subsets of SSc. An inflammatory gene module from CD16+ monocytes, which included KLF10, PLAUR, JUNB and JUND, showed the greatest discrimination between SSc and HC. One of the clusters of SSc monocytes identified by single-cell RNA-seq analysis characteristically expressed these inflammatory co-expressed genes and was similar to lung infiltrating FCN1hi monocytes expressing IL1B. CONCLUSIONS: Our integrated analysis of bulk and single cell RNA-seq analysis identified an inflammatory gene module and a cluster of monocytes that are relevant to SSc pathophysiology. They could serve as candidate novel therapeutic targets in SSc.


Assuntos
Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Monócitos/metabolismo , RNA-Seq/métodos , Escleroderma Sistêmico/genética , Análise de Célula Única/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Análise por Conglomerados , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/classificação , Monócitos/citologia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Escleroderma Sistêmico/terapia
11.
J Autoimmun ; 119: 102617, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33677398

RESUMO

OBJECTIVE: Previous gene expression analyses seeking genes specific to antineutrophil cytoplasmic antibody-associated vasculitis (AAV) have been limited due to crude cell separation and the use of microarrays. This study aims to identify AAV-specific gene expression profiles in a way that overcomes those limitations. METHODS: Blood samples were collected from 26 AAV patients and 28 healthy controls (HCs). Neutrophils were isolated by negative selection, whereas 19 subsets of peripheral blood mononuclear cells were sorted by fluorescence assisted cell sorting. RNA-sequencing was then conducted for each sample, and iterative weighted gene correlation network analysis (iterativeWGCNA) and random forest were consecutively applied to identify the most influential gene module in distinguishing AAV from HCs. Correlations of the identified module with clinical parameters were evaluated, and the biological role was assessed with hub gene identification and pathway analysis. Particularly, the module's association with neutrophil extracellular trap formation, NETosis, was analyzed. Finally, the module's overlap with GWAS-identified autoimmune disease genes (GADGs) was assessed for validation. RESULTS: A neutrophil module (Neu_M20) was ranked top in the random forest analysis among 255 modules created by iterativeWGCNA. Neu_M20 correlated with disease activity and neutrophil counts but not with the presence of antineutrophil cytoplasmic antibody. The module comprised pro-inflammatory genes, including those related to NETosis, supported by experimental evidence. The genes in the module significantly overlapped GADGs. CONCLUSION: We identified the distinct group of pro-inflammatory genes in neutrophils, which characterize AAV. Further investigations are warranted to confirm our findings as they could serve as novel therapeutic targets.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/diagnóstico , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/etiologia , Biomarcadores , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Transcriptoma , Anticorpos Anticitoplasma de Neutrófilos/efeitos adversos , Anticorpos Anticitoplasma de Neutrófilos/imunologia , Estudos de Casos e Controles , Biologia Computacional , Armadilhas Extracelulares/imunologia , Armadilhas Extracelulares/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunofenotipagem , Neutrófilos/imunologia , Neutrófilos/metabolismo
12.
Proc Natl Acad Sci U S A ; 113(50): E8131-E8140, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911796

RESUMO

Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease characterized by multiorgan inflammation induced by autoantibodies. Early growth response gene 2 (Egr2), a transcription factor essential for T-cell anergy induction, controls systemic autoimmunity in mice and humans. We have previously identified a subpopulation of CD4+ regulatory T cells, CD4+CD25-LAG3+ cells, that characteristically express both Egr2 and LAG3 and control mice model of lupus via TGF-ß3 production. However, due to the mild phenotype of lymphocyte-specific Egr2-deficient mice, the presence of an additional regulator has been speculated. Here, we show that Egr2 and Egr3 expressed in T cells cooperatively prevent humoral immune responses by supporting TGF-ß3 secretion. T cell-specific Egr2/Egr3 double-deficient (Egr2/3DKO) mice spontaneously developed an early onset lupus-like disease that was more severe than in T cell-specific Egr2-deficient mice. In accordance with the observation that CD4+CD25-LAG3+ cells from Egr2/3DKO mice completely lost the capacity to produce TGF-ß3, the excessive germinal center reaction in Egr2/3DKO mice was suppressed by the adoptive transfer of WT CD4+CD25-LAG3+ cells or treatment with a TGF-ß3-expressing vector. Intriguingly, latent TGF-ß binding protein (Ltbp)3 expression maintained by Egr2 and Egr3 was required for TGF-ß3 production from CD4+CD25-LAG3+ cells. Because Egr2 and Egr3 did not demonstrate cell intrinsic suppression of the development of follicular helper T cells, Egr2- and Egr3-dependent TGF-ß3 production by CD4+CD25-LAG3+ cells is critical for controlling excessive B-cell responses. The unique attributes of Egr2/Egr3 in T cells may provide an opportunity for developing novel therapeutics for autoantibody-mediated diseases including SLE.


Assuntos
Autoimunidade , Proteína 2 de Resposta de Crescimento Precoce/imunologia , Proteína 3 de Resposta de Crescimento Precoce/imunologia , Proteínas de Ligação a TGF-beta Latente/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta3/biossíntese , Imunidade Adaptativa , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Diferenciação Celular , Modelos Animais de Doenças , Proteína 2 de Resposta de Crescimento Precoce/deficiência , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 3 de Resposta de Crescimento Precoce/deficiência , Proteína 3 de Resposta de Crescimento Precoce/genética , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Centro Germinativo/patologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia , Linfócitos T Reguladores/patologia
13.
Molecules ; 24(3)2019 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-30696004

RESUMO

Highly efficient, operationally stable, and pure-color organic light-emitting diodes (OLEDs) are of considerable significance for developing practical wide-color-gamut displays. Further, we have demonstrated the feasibility of an efficient pure green phosphorescent OLED (PHOLED) by employing a narrow-band platinum complex and a top-emitting structure. The utilization of the thermally activated delayed fluorescence (TADF) material as the phosphorescent host is expected to serve as a promising solution for obtaining operationally stable PHOLEDs with high color purity. However, the emission spectrum of the platinum complex in the TADF host exhibits a considerably broad emission spectrum. This study investigates the cause of the spectral change by evaluating the photoluminescence spectra of the platinum complex in various hosts exhibiting different molecular structures. The triazine unit in the host material was observed to result in exciplex formation between the lowest unoccupied molecular orbital (LUMO) of the host and the highest occupied molecular orbital (HOMO) of the platinum complex. Therefore, the TADF material that sterically hinders the triazine unit is considered to be suitable to prevent both exciplex formation and spectral broadening.


Assuntos
Medições Luminescentes , Compostos Organoplatínicos/química , Platina/química , Análise Espectral , Carbazóis/química , Medições Luminescentes/métodos , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Compostos Organoplatínicos/análise , Platina/análise , Análise Espectral/métodos
14.
Ann Rheum Dis ; 77(4): 602-611, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29331962

RESUMO

OBJECTIVES: Idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of rare autoimmune diseases in which both genetic and environmental factors play important roles. To identify genetic factors of IIM including polymyositis, dermatomyositis (DM) and clinically amyopathic DM (CADM), we performed the first genome-wide association study for IIM in an Asian population. METHODS: We genotyped and tested 496 819 single nucleotide polymorphism for association using 576 patients with IIM and 6270 control subjects. We also examined the causal mechanism of disease-associated variants by in silico analyses using publicly available data sets as well as by in in vitro analyses using reporter assays and apoptosis assays. RESULTS: We identified a variant in WDFY4 that was significantly associated with CADM (rs7919656; OR=3.87; P=1.5×10-8). This variant had a cis-splicing quantitative trait locus (QTL) effect for a truncated WDFY4isoform (tr-WDFY4), with higher expression in the risk allele. Transexpression QTL analysis of this variant showed a positive correlation with the expression of NF-κB associated genes. Furthermore, we demonstrated that both WDFY4 and tr-WDFY4 interacted with pattern recognition receptors such as TLR3, TLR4, TLR9 and MDA5 and augmented the NF-κB activation by these receptors. WDFY4 isoforms also enhanced MDA5-induced apoptosis to a greater extent in the tr-WDFY4-transfected cells. CONCLUSIONS: As CADM is characterised by the appearance of anti-MDA5 autoantibodies and severe lung inflammation, the WDFY4 variant may play a critical role in the pathogenesis of CADM.


Assuntos
Dermatomiosite/genética , Helicase IFIH1 Induzida por Interferon/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Splicing de RNA/genética , Transdução de Sinais/genética , Adulto , Idoso , Alelos , Apoptose/genética , Povo Asiático/genética , Autoanticorpos/genética , Estudos de Casos e Controles , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Técnicas de Genotipagem , Humanos , Helicase IFIH1 Induzida por Interferon/imunologia , Masculino , Pessoa de Meia-Idade , NF-kappa B/genética , Polimorfismo de Nucleotídeo Único , Polimiosite/genética , Isoformas de Proteínas/genética , Locos de Características Quantitativas/genética , Fatores de Risco
15.
Mod Rheumatol ; 28(4): 621-625, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29057700

RESUMO

OBJECTIVES: The characteristics of lymphoproliferative disorders (LPD) in patients with rheumatoid arthritis (RA) remain unclear. Therefore, we retrospectively analyzed the clinical characteristics of these patients in our department. METHODS: Twenty RA patients who developed LPD between April 2003 and August 2016 in our department were analyzed. RESULTS: All of the RA patients who developed LPD had been treated with methotrexate (MTX). The median weekly and total dosages of MTX were 6.8 mg/week and 2530 mg, respectively. The median duration of MTX administration was eight years. Nineteen patients (95%) achieved complete remission (CR) and 15 (75%) achieved CR with MTX cessation alone. Based on the pathological findings, we divided MTX-associated LPD patients into two groups (n = 16); polymorphic LPD (31%) and other groups. CR with MTX cessation alone was achieved in 5 (100%) and 6 (54.5%) patients in the polymorphic LPD and other groups, respectively (p = .12). Moreover, the duration from the cessation of MTX to CR was significantly shorter in the polymorphic LPD group than in the other group (5.3 months vs 12.6 months, p = .01, respectively). CONCLUSION: Polymorphic LPD, which was the most frequent pathological diagnosis in this cohort, was associated with a higher incidence of CR and a significantly shorter duration to CR.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Transtornos Linfoproliferativos/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Antirreumáticos/uso terapêutico , Artrite Reumatoide/complicações , Estudos de Coortes , Feminino , Humanos , Incidência , Masculino , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Indução de Remissão
16.
Mod Rheumatol ; 27(1): 22-28, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27142966

RESUMO

OBJECTIVES: Acute or subacute exacerbations are recognized as a severe complication of rheumatoid arthritis-associated interstitial lung disease (RA-ILD). Nevertheless, the role of intensive immunosuppression in RA-ILD remains elusive. We attempted to evaluate the clinical characteristics and efficacy of immunosuppressive treatment in exacerbated RA-ILD. METHODS: Clinical data, including respiratory function, imaging, treatment, and prognosis, were retrospectively collected for 17 patients with RA-ILD who required hospitalization at the University of Tokyo Hospital due to an acute exacerbation (12 patients) or subacute exacerbation (5 patients). RESULTS: Patients with RA-ILD demonstrated a significantly higher titers of anticyclic citrullinated peptide antibodies compared with RA patients in Japanese Ninja registry, suggesting the role of adaptive immunity. Immunosuppressive treatment suppressed the deterioration of pulmonary functions with improved ground grass opacity and consolidation. In particular, in patients with less fibrosis on computed tomography (CT) images showed a better response to treatment. Although five patients treated with combination therapy, including cyclophosphamide, showed a severely decreased lung volume, these intensive therapies provided a good prognosis without fatalities for the average observation period of 474 days. CONCLUSIONS: Immunosuppressive therapy is effective for exacerbations of RA-ILD. For severe cases with low respiratory function, intensive therapy, including cyclophosphamide, has a potential to improve the prognosis.


Assuntos
Artrite Reumatoide , Ciclofosfamida/uso terapêutico , Doenças Pulmonares Intersticiais , Pulmão , Peptídeos Cíclicos/imunologia , Idoso , Artrite Reumatoide/complicações , Artrite Reumatoide/imunologia , Autoanticorpos/sangue , Progressão da Doença , Feminino , Hospitalização/estatística & dados numéricos , Humanos , Imunossupressores/uso terapêutico , Japão , Pulmão/diagnóstico por imagem , Pulmão/fisiopatologia , Doenças Pulmonares Intersticiais/etiologia , Doenças Pulmonares Intersticiais/imunologia , Doenças Pulmonares Intersticiais/fisiopatologia , Doenças Pulmonares Intersticiais/terapia , Masculino , Pessoa de Meia-Idade , Prognóstico , Testes de Função Respiratória/métodos , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/métodos , Resultado do Tratamento
17.
Rheumatology (Oxford) ; 55(suppl 2): ii76-ii81, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27856665

RESUMO

SLE is an autoimmune disease characterized by multiple organ damage mediated by autoantibodies and autoreactive T cells. Approaches utilizing genetically engineered mice as well as genome-wide association studies have identified a number of lupus-related genes. Recently, early growth response gene 2 (Egr2) and Egr3 have emerged as regulatory molecules that suppress excessive immune responses. Mice deficient for Egr2 and Egr3 develop a lupus-like disease with dysregulated activation of effector T cells. Furthermore, Egr2 and Egr3 confer suppressive activity to CD4+ T cells and regulate the production of inhibitory cytokines such as IL-10 and TGF-ß1. These findings may have implications for a wide range of immune-related pathologies and suggest the possibility that efforts exploiting Egr2 and Egr3 could aid in the development of therapeutic applications. This review summarizes the recent advances regarding the roles of Egr2 and Egr3 on T cells in the control of autoimmunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proteína 2 de Resposta de Crescimento Precoce/imunologia , Proteína 3 de Resposta de Crescimento Precoce/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 3 de Resposta de Crescimento Precoce/genética , Humanos , Interleucina-10/imunologia , Interleucina-17/imunologia , Lúpus Eritematoso Sistêmico/genética , Camundongos , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/imunologia
18.
J Immunol ; 193(4): 1728-36, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25024389

RESUMO

Dendritic cells (DCs) are proficient APCs that play crucial roles in the immune responses to various Ags and pathogens and polarize Th cell immune responses. Lnk/SH2B adaptor protein 3 (Sh2b3) is an intracellular adaptor protein that regulates B lymphopoiesis, megakaryopoiesis, and expansion of hematopoietic stem cells by constraining cytokine signals. Recent genome-wide association studies have revealed a link between polymorphism in this adaptor protein and autoimmune diseases, including type 1 diabetes and celiac disease. We found that Lnk/Sh2b3 was also expressed in DCs and investigated its role in the production and function of DC lineage cells. In Lnk(-/-) mice, DC numbers were increased in the spleen and lymph nodes, and growth responses of bone marrow-derived DCs to GM-CSF were augmented. Mature DCs from Lnk(-/-) mice were hypersensitive and showed enhanced responses to IL-15 and GM-CSF. Compared to normal DCs, Lnk(-/-) DCs had enhanced abilities to support the differentiation of IFN-γ-producing Th1 cells from naive CD4(+) T cells. This was due to their elevated expression of IL-12Rß1 and increased production of IFN-γ. Lnk(-/-) DCs supported the appearance of IFN-γ-producing T cells even under conditions in which normal DCs supported induction of regulatory T cells. These results indicated that Lnk/Sh2b3 plays a regulatory role in the expansion of DCs and might influence inflammatory immune responses in peripheral lymphoid tissues.


Assuntos
Células Dendríticas/imunologia , Interferon gama/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Linfopoese/imunologia , Células Th1/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Adjuvantes Imunológicos/farmacologia , Animais , Células da Medula Óssea/citologia , Doença Celíaca/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Estudo de Associação Genômica Ampla , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/imunologia , Inflamação/imunologia , Interleucina-12/farmacologia , Interleucina-15/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Linfonodos/citologia , Ativação Linfocitária/imunologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , Receptores de Interleucina-12/biossíntese , Baço/citologia , Linfócitos T Reguladores/imunologia , Trombopoese/imunologia
19.
Nihon Rinsho ; 74(6): 889-96, 2016 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-27311174

RESUMO

Rheumatoid arthritis (RA) is a common autoimmune disease with a prevalence of 0.5-1.0% worldwide. Although advances in understanding the pathogenesis of RA have led to new therapeutics with good outcomes, the real cause of the disease is still unknown. RA is characterized by synovial inflammation and hyperplasia, which erodes cartilage and bone, and autoantibody production (rheumatoid factor (RF) and anti-citrullinated protein antibody (ACPA)). There are many critical questions on the mechanism of the disease onset and progression: How genetic and environmental factors interact with each other? Why does the inflammatory response localize in joints? What are the key players to perpetuate synovial inflammation? In this review, we summarize pathogenetic advances in these issues especially from the point of view of basic research.


Assuntos
Artrite Reumatoide , Animais , Formação de Anticorpos/imunologia , Artrite Reumatoide/etiologia , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Autoanticorpos , Diferenciação Celular , Quimiocinas/fisiologia , Progressão da Doença , Fibroblastos , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Cadeias HLA-DRB1 , Humanos , Inflamação/imunologia , Inflamação/patologia , Articulações/imunologia , Leucócitos/imunologia , Camundongos , Peptídeos Cíclicos/imunologia , Prevotella , Fator Reumatoide , Fumar/efeitos adversos , Membrana Sinovial/citologia , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Células Th17/citologia
20.
Eur J Immunol ; 44(6): 1622-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24536025

RESUMO

The intracellular adaptor Lnk (also known as SH2B3) regulates cytokine signals that control lymphohematopoiesis, and Lnk(-/-) mice have expanded B-cell, megakaryocyte, and hematopoietic stem-cell populations. Moreover, mutations in the LNK gene are found in patients with myeloproliferative disease, whereas LNK polymorphisms have recently been associated with inflammatory and autoimmune diseases, including celiac disease. Here, we describe a previously unrecognized function of Lnk in the control of inflammatory CD8(+) T-cell proliferation and in intestinal homeostasis. Mature T cells from newly generated Lnk-Venus reporter mice had low but substantial expression of Lnk, whereas Lnk expression was downregulated during homeostatic T-cell proliferation under lymphopenic conditions. The numbers of CD44(hi) IFN-γ(+) CD8(+) effector or memory T cells were found to be increased in Lnk(-/-) mice, which also exhibited shortening of villi in the small intestine. Lnk(-/-) CD8(+) T cells survived longer in response to stimulation with IL-15 and proliferated even in nonlymphopenic hosts. Transfer of Lnk(-/-) CD8(+) T cells together with WT CD4(+) T cells into Rag2-deficient mice recapitulated a sign of villous abnormality. Our results reveal a link between Lnk and immune cell-mediated intestinal tissue destruction.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Homeostase/imunologia , Intestinos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos T CD8-Positivos/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Homeostase/genética , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Polimorfismo Genético/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA