Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int Immunol ; 36(3): 129-139, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38041796

RESUMO

To meet the energetic requirements associated with activation, proliferation, and survival, T cells switch their metabolic signatures from energetically quiescent to activated. However, little is known about the role of metabolic pathway controlling the development of invariant natural killer T (iNKT) cells. In the present study, we found that acetyl-CoA carboxylase 1 (ACC1), a rate-limiting enzyme for the fatty acid biosynthesis pathway, plays an essential role in the development of iNKT cells in the thymus. Mice lacking T-cell specific ACC1 showed a reduced number of iNKT cells with an increased proportion of iNKT cells at immature stages 0 and 1. Furthermore, mixed bone marrow (BM) chimera experiments revealed that T-cell intrinsic ACC1 expression was selectively important for the development of thymic iNKT cells, especially for the differentiation of the NKT1 cell subset. Our single-cell RNA-sequencing (scRNA-seq) data and functional analysis demonstrated that ACC1 is responsible for survival of developing iNKT cells. Thus, these findings highlighted a novel role of ACC1 in controlling thymic iNKT cell development mediated by the control of cell survival.


Assuntos
Células T Matadoras Naturais , Camundongos , Animais , Timo , Diferenciação Celular , Adipogenia , Ácidos Graxos/metabolismo , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo
2.
Immunity ; 42(2): 294-308, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25692703

RESUMO

Memory CD4(+) T helper (Th) cells provide long-term protection against pathogens and are essential for the development of vaccines; however, some antigen-specific memory Th cells also drive immune-related pathology, including asthma. The mechanisms regulating the pathogenicity of memory Th cells remain poorly understood. We found that interleukin-33 (IL-33)-ST2 signals selectively licensed memory Th2 cells to induce allergic airway inflammation via production of IL-5 and that the p38 MAP kinase pathway was a central downstream target of IL-33-ST2 in memory Th2 cells. In addition, we found that IL-33 induced upregulation of IL-5 by memory CD4(+) T cells isolated from nasal polyps of patients with eosinophilic chronic rhinosinusitis. Thus, IL-33-ST2-p38 signaling appears to directly instruct pathogenic memory Th2 cells to produce IL-5 and induce eosinophilic inflammation.


Assuntos
Asma/imunologia , Interleucina-5/imunologia , Interleucinas/imunologia , Receptores de Interleucina/imunologia , Células Th2/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Animais , Asma/patologia , Células Cultivadas , Humanos , Memória Imunológica/imunologia , Inflamação/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Interleucina-5/biossíntese , Interleucinas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pólipos Nasais/imunologia , Eosinofilia Pulmonar/imunologia , Interferência de RNA , RNA Interferente Pequeno , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Interleucina/genética , Sinusite/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética
3.
Commun Biol ; 7(1): 622, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783005

RESUMO

Recent studies have highlighted the significance of cellular metabolism in the initiation of clonal expansion and effector differentiation of T cells. Upon exposure to antigens, naïve CD4+ T cells undergo metabolic reprogramming to meet their metabolic requirements. However, only few studies have simultaneously evaluated the changes in protein and metabolite levels during T cell differentiation. Our research seeks to fill the gap by conducting a comprehensive analysis of changes in levels of metabolites, including sugars, amino acids, intermediates of the TCA cycle, fatty acids, and lipids. By integrating metabolomics and proteomics data, we discovered that the quantity and composition of cellular lipids underwent significant changes in different effector Th cell subsets. Especially, we found that the sphingolipid biosynthesis pathway was commonly activated in Th1, Th2, Th17, and iTreg cells and that inhibition of this pathway led to the suppression of Th17 and iTreg cells differentiation. Additionally, we discovered that Th17 and iTreg cells enhance glycosphingolipid metabolism, and inhibition of this pathway also results in the suppression of Th17 and iTreg cell generation. These findings demonstrate that the utility of our combined metabolomics and proteomics analysis in furthering the understanding of metabolic transition during Th cell differentiation.


Assuntos
Diferenciação Celular , Metabolômica , Proteômica , Esfingolipídeos , Esfingolipídeos/metabolismo , Esfingolipídeos/biossíntese , Proteômica/métodos , Animais , Metabolômica/métodos , Camundongos , Camundongos Endogâmicos C57BL
4.
Sci Immunol ; 8(86): eadd4346, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37540735

RESUMO

Metabolic fluxes involving fatty acid biosynthesis play essential roles in controlling the differentiation of T helper 17 (TH17) cells. However, the exact enzymes and lipid metabolites involved, as well as their link to promoting the core gene transcriptional signature required for the differentiation of TH17 cells, remain largely unknown. From a pooled CRISPR-based screen and unbiased lipidomics analyses, we identified that 1-oleoyl-lysophosphatidylethanolamine could act as a lipid modulator of retinoid-related orphan receptor gamma t (RORγt) activity in TH17 cells. In addition, we specified five enzymes, including Gpam, Gpat3, Lplat1, Pla2g12a, and Scd2, suggestive of the requirement of glycerophospholipids with monounsaturated fatty acids being required for the transcription of Il17a. 1-Oleoyl-lysophosphatidylethanolamine was reduced in Pla2g12a-deficient TH17 cells, leading to the abolition of interleukin-17 (IL-17) production and disruption to the core transcriptional program required for the differentiation of TH17 cells. Furthermore, mice with T cell-specific deficiency of Pla2g12a failed to develop disease in an experimental autoimmune encephalomyelitis model of multiple sclerosis. Thus, our data indicate that 1-oleoyl-lysophosphatidylethanolamine is a lipid metabolite that promotes RORγt-induced TH17 cell differentiation and the pathogenicity of TH17 cells.


Assuntos
Encefalomielite Autoimune Experimental , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Camundongos , Animais , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Diferenciação Celular , Lipídeos
5.
DNA Res ; 30(1)2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36579714

RESUMO

Functionally distinct CD4+ helper T (Th) cell subsets, including Th1, Th2, Th17, and regulatory T cells (Treg), play a pivotal role in the regulation of acquired immunity. Although the key proteins involved in the regulation of Th cell differentiation have already been identified how the proteogenomic landscape changes during the Th cell activation remains unclear. To address this issue, we characterized proteogenomic signatures of differentiation to each Th cell subsets by RNA sequencing and liquid chromatography-assisted mass spectrometry, which enabled us to simultaneously quantify more than 10,000 protein-coding transcripts and 8,000 proteins in a single-shot. The results indicated that T cell receptor activation affected almost half of the transcript and protein levels in a low correlative and gene-specific manner, and specific cytokine treatments modified the transcript and protein profiles in a manner specific to each Th cell subsets: Th17 and Tregs particularly exhibited unique proteogenomic signatures compared to other Th cell subsets. Interestingly, the in-depth proteome data revealed that mRNA profiles alone were not enough to delineate functional changes during Th cell activation, suggesting that the proteogenomic dataset obtained in this study serves as a unique and indispensable data resource for understanding the comprehensive molecular mechanisms underlying effector Th cell differentiation.


Assuntos
Linfócitos T CD4-Positivos , Proteogenômica , Linfócitos T CD4-Positivos/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Diferenciação Celular/genética
6.
Front Immunol ; 13: 904875, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36059459

RESUMO

Type I interferons (type I-IFN) are critical for the host defense to viral infection, and at the same time, the dysregulation of type I-IFN responses leads to autoinflammation or autoimmunity. Recently, we reported that the decrease in monounsaturated fatty acid caused by the genetic deletion of Scd2 is essential for the activation of type I-IFN signaling in CD4+ Th1 cells. Although interferon regulatory factor (IRF) is a family of homologous proteins that control the transcription of type I-IFN and interferon stimulated genes (ISGs), the member of the IRF family that is responsible for the type I-IFN responses induced by targeting of SCD2 remains unclear. Here, we report that the deletion of Scd2 triggered IRF3 activation for type I-IFN production, resulting in the nuclear translocation of IRF9 to induce ISG transcriptome in Th1 cells. These data led us to hypothesize that IRF9 plays an essential role in the transcriptional regulation of ISGs in Scd2-deleted (sgScd2) Th1 cells. By employing ChIP-seq analyses, we found a substantial percentage of the IRF9 target genes were shared by sgScd2 and IFNß-treated Th1 cells. Importantly, our detailed analyses identify a unique feature of IRF9 binding in sgScd2 Th1 cells that were not observed in IFNß-treated Th1 cells. In addition, our combined analyses of transcriptome and IRF9 ChIP-seq revealed that the autoimmunity related genes, which increase in patient with SLE, were selectively increased in sgScd2 Th1 cells. Thus, our findings provide novel mechanistic insights into the process of fatty acid metabolism that is essential for the type I-IFN response and the activation of the IRF family in CD4+ T cells.


Assuntos
Linfócitos T CD4-Positivos , Fatores Reguladores de Interferon , Interferon Tipo I , Estearoil-CoA Dessaturase , Antivirais , Linfócitos T CD4-Positivos/metabolismo , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interferon Tipo I/metabolismo , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Estearoil-CoA Dessaturase/genética , Transcriptoma
7.
J Exp Med ; 218(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34813654

RESUMO

T cells possess distinguishing effector functions and drive inflammatory disorders. We have previously identified IL-5-producing Th2 cells as the pathogenic population predominantly involved in the pathology of allergic inflammation. However, the cell-intrinsic signaling pathways that control the pathogenic Th2 cell function are still unclear. We herein report the high expression of acetyl-CoA carboxylase 1 (ACC1) in the pathogenic CD4+ T cell population in the lung and skin. The genetic deletion of CD4+ T cell-intrinsic ACC1 dampened eosinophilic and basophilic inflammation in the lung and skin by constraining IL-5 or IL-3 production. Mechanistically, ACC1-dependent fatty acid biosynthesis induces the pathogenic cytokine production of CD4+ T cells via metabolic reprogramming and the availability of acetyl-CoA for epigenetic regulation. We thus identified a distinct phenotype of the pathogenic T cell population in the lung and skin, and ACC1 was shown to be an essential regulator controlling the pathogenic function of these populations to promote type 2 inflammation.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Toxidermias/patologia , Pneumonia/patologia , Células Th2/patologia , Acetil-CoA Carboxilase/genética , Administração Tópica , Animais , Basófilos/metabolismo , Basófilos/patologia , Linfócitos T CD4-Positivos/patologia , Calcitriol/análogos & derivados , Calcitriol/toxicidade , Toxidermias/tratamento farmacológico , Toxidermias/genética , Toxidermias/metabolismo , Ácidos Graxos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-3/metabolismo , Interleucina-5/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Pneumonia/genética , Pneumonia/metabolismo , Células Th2/metabolismo
8.
Cell Rep ; 37(6): 109921, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34758300

RESUMO

Regulatory T (Treg) cells are critical for immunological tolerance and immune homeostasis. Treg cells strongly rely on mitochondrial metabolism and show a lower level of glycolysis. However, little is known about the role of lipid metabolism in the regulation of Treg cell homeostasis. Some members of the ACSL family of acyl-coenzyme A (CoA) synthases are expressed in T cells, but their function remains unclear. A combination of RNA-sequencing and proteome analyses shows that Acsbg1, a member of ACSL, is selectively expressed in Treg cells. We show that the genetic deletion of Acsbg1 not only causes mitochondrial dysfunction, but it also dampens other metabolic pathways. The extrinsic supplementation of Acsbg1-deficient Treg cells with oleoyl-CoA restores the phenotype of the Treg metabolic signature. Furthermore, this pathway in ST2+ effector Treg cells enhances immunosuppressive capacity in airway inflammation. Thus, Acsbg1 serves as a metabolic checkpoint governing Treg cell homeostasis and the resolution of lung inflammation.


Assuntos
Coenzima A Ligases/metabolismo , Metabolismo Energético , Pulmão/enzimologia , Mitocôndrias/enzimologia , Pneumonia/enzimologia , Linfócitos T Reguladores/enzimologia , Animais , Coenzima A Ligases/genética , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Regulação Enzimológica da Expressão Gênica , Homeostase , Interleucina-33 , Pulmão/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/imunologia , Biogênese de Organelas , Pneumonia/genética , Pneumonia/imunologia , Transdução de Sinais , Linfócitos T Reguladores/imunologia
9.
Commun Biol ; 4(1): 820, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34188173

RESUMO

Host lipid metabolism and viral responses are intimately connected. However, the process by which the acquired immune systems adapts lipid metabolism to meet demands, and whether or not the metabolic rewiring confers a selective advantage to host immunity, remains unclear. Here we show that viral infection attenuates the expression of genes related to lipid metabolism in murine CD4+ T cells, which in turn increases the expression of antiviral genes. Inhibition of the fatty acid synthesis pathway substantially increases the basal expression of antiviral genes via the spontaneous production of type I interferon (IFN). Using a combination of CRISPR/Cas9-mediated genome editing technology and a global lipidomics analysis, we found that the decrease in monounsaturated fatty acid caused by genetic deletion of Scd2 in mice was crucial for the induction of an antiviral response through activation of the cGAS-STING pathway. These findings demonstrate the important relationship between fatty acid biosynthesis and type I IFN responses that enhances the antiviral response.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Ácidos Graxos Monoinsaturados/metabolismo , Interferon Tipo I/farmacologia , Proteínas de Membrana/fisiologia , Nucleotidiltransferases/fisiologia , Estearoil-CoA Dessaturase/fisiologia , Viroses/imunologia , Animais , Interações Hospedeiro-Patógeno , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transdução de Sinais , Viroses/metabolismo
10.
Nat Metab ; 1(2): 261-275, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-32694782

RESUMO

Immunological memory is central to adaptive immunity and protection from disease. Changing metabolic demands as antigen-specific T cells transition from effector to memory cells have been well documented, but the cell-specific pathways and molecules that govern this transition are poorly defined. Here we show that genetic deletion of ACC1, a rate-limiting enzyme in fatty acid biosynthesis, enhances the formation of CD4+ T memory cells. ACC1-deficient effector helper T (Th) cells have similar metabolic signatures to wild-type memory Th cells, and expression of the gene encoding ACC1, Acaca, was inversely correlated with a memory gene signature in individual cells. Inhibition of ACC1 function enhances memory T cell formation during parasite infection in mice. Using single-cell analyses we identify a memory precursor-enriched population (CCR7hiCD137lo) present during early differentiation of effector CD4+ T cells. Our data indicate that fatty acid metabolism directs cell fate determination during the generation of memory CD4+ T cells.


Assuntos
Acetil-CoA Carboxilase/fisiologia , Linfócitos T CD4-Positivos/imunologia , Ácidos Graxos/biossíntese , Memória Imunológica/fisiologia , Animais , Linhagem da Célula , Camundongos , Camundongos Endogâmicos BALB C
11.
Nat Commun ; 9(1): 4231, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30315197

RESUMO

ST2hi memory-type Th2 cells are identified as a pathogenic subpopulation in eosinophilic airway inflammation. These ST2hi pathogenic Th2 cells produce large amount of IL-5 upon T cell receptor stimulation, but not in response to IL-33 treatment. By contrast, IL-33 alone induces cytokine production in ST2+ group 2 innate lymphoid cells (ILC2). Here we show that a MAPK phosphatase Dusp10 is a key negative regulator of IL-33-induced cytokine production in Th2 cells. In this regard, Dusp10 is expressed by ST2hi pathogenic Th2 cells but not by ILC2, and Dusp10 expression inhibits IL-33-induced cytokine production. Mechanistically, this inhibition is mediated by DUSP10-mediated dephosphorylation and inactivation of p38 MAPK, resulting in reduced GATA3 activity. The deletion of Dusp10 renders ST2hi Th2 cells capable of producing IL-5 by IL-33 stimulation. Our data thus suggest that DUSP10 restricts IL-33-induced cytokine production in ST2hi pathogenic Th2 cells by controlling p38-GATA3 activity.


Assuntos
Citocinas/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Interleucina-33/farmacologia , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Animais , Animais Geneticamente Modificados , Imunoprecipitação da Cromatina , Fosfatases de Especificidade Dupla/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fator de Transcrição GATA3/metabolismo , Humanos , Immunoblotting , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Nat Commun ; 7: 13683, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27901044

RESUMO

To fulfil the bioenergetic requirements for increased cell size and clonal expansion, activated T cells reprogramme their metabolic signatures from energetically quiescent to activated. However, the molecular mechanisms and essential components controlling metabolic reprogramming in T cells are not well understood. Here, we show that the mTORC1-PPARγ pathway is crucial for the fatty acid uptake programme in activated CD4+ T cells. This pathway is required for full activation and rapid proliferation of naive and memory CD4+ T cells. PPARγ directly binds and induces genes associated with fatty acid uptake in CD4+ T cells in both mice and humans. The PPARγ-dependent fatty acid uptake programme is critical for metabolic reprogramming. Thus, we provide important mechanistic insights into the metabolic reprogramming mechanisms that govern the expression of key enzymes, fatty acid metabolism and the acquisition of an activated phenotype during CD4+ T cell activation.


Assuntos
Ácidos Graxos/metabolismo , Ativação Linfocitária/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , PPAR gama/metabolismo , Transdução de Sinais/fisiologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Proliferação de Células/fisiologia , Imunoprecipitação da Cromatina , Técnicas de Silenciamento de Genes , Voluntários Saudáveis , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , PPAR gama/antagonistas & inibidores , PPAR gama/genética , RNA Interferente Pequeno/metabolismo , Serina-Treonina Quinases TOR , Regulação para Cima/fisiologia
13.
Cell Rep ; 12(6): 1042-55, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26235623

RESUMO

Chronic inflammation due to obesity contributes to the development of metabolic diseases, autoimmune diseases, and cancer. Reciprocal interactions between metabolic systems and immune cells have pivotal roles in the pathogenesis of obesity-associated diseases, although the mechanisms regulating obesity-associated inflammatory diseases are still unclear. In the present study, we performed transcriptional profiling of memory phenotype CD4 T cells in high-fat-fed mice and identified acetyl-CoA carboxylase 1 (ACC1, the gene product of Acaca) as an essential regulator of Th17 cell differentiation in vitro and of the pathogenicity of Th17 cells in vivo. ACC1 modulates the DNA binding of RORγt to target genes in differentiating Th17 cells. In addition, we found a strong correlation between IL-17A-producing CD45RO(+)CD4 T cells and the expression of ACACA in obese subjects. Thus, ACC1 confers the appropriate function of RORγt through fatty acid synthesis and regulates the obesity-related pathology of Th17 cells.


Assuntos
Acetiltransferases/metabolismo , Diferenciação Celular/fisiologia , Obesidade/metabolismo , Células Th17/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Antígenos Comuns de Leucócito/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa