Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45.503
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nat Immunol ; 18(9): 1035-1045, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28759003

RESUMO

MLL4 is an essential subunit of the histone H3 Lys4 (H3K4)-methylation complexes. We found that MLL4 deficiency compromised the development of regulatory T cells (Treg cells) and resulted in a substantial decrease in monomethylated H3K4 (H3K4me1) and chromatin interaction at putative gene enhancers, a considerable portion of which were not direct targets of MLL4 but were enhancers that interacted with MLL4-bound sites. The decrease in H3K4me1 and chromatin interaction at the enhancers not bound by MLL4 correlated with MLL4 binding at distant interacting regions. Deletion of an upstream MLL4-binding site diminished the abundance of H3K4me1 at the regulatory elements of the gene encoding the transcription factor Foxp3 that were looped to the MLL4-binding site and compromised both the thymic differentiation and the inducible differentiation of Treg cells. We found that MLL4 catalyzed methylation of H3K4 at distant unbound enhancers via chromatin looping, which identifies a previously unknown mechanism for regulating the T cell enhancer landscape and affecting Treg cell differentiation.


Assuntos
Diferenciação Celular/genética , Cromatina/metabolismo , Fatores de Transcrição Forkhead/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Linfócitos T Reguladores , Animais , Sistemas CRISPR-Cas , Citocinas/imunologia , Citometria de Fluxo , Regulação da Expressão Gênica , Immunoblotting , Técnicas In Vitro , Metilação , Camundongos
2.
Nat Immunol ; 18(11): 1197-1206, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28920951

RESUMO

Activated natural killer (NK) cells engage in a robust metabolic response that is required for normal effector function. Using genetic, pharmacological and metabolic analyses, we demonstrated an essential role for Srebp transcription factors in cytokine-induced metabolic reprogramming of NK cells that was independent of their conventional role in the control of lipid synthesis. Srebp was required for elevated glycolysis and oxidative phosphorylation and promoted a distinct metabolic pathway configuration in which glucose was metabolized to cytosolic citrate via the citrate-malate shuttle. Preventing the activation of Srebp or direct inhibition of the citrate-malate shuttle inhibited production of interferon-γ and NK cell cytotoxicity. Thus, Srebp controls glucose metabolism in NK cells, and this Srebp-dependent regulation is critical for NK cell effector function.


Assuntos
Glucose/metabolismo , Glicólise , Células Matadoras Naturais/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Animais , Proliferação de Células , Citocinas/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Células Matadoras Naturais/imunologia , Lipídeos/biossíntese , Fosforilação Oxidativa , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
3.
Nat Immunol ; 18(7): 780-790, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28553951

RESUMO

The acquisition of a protective vertebrate immune system hinges on the efficient generation of a diverse but self-tolerant repertoire of T cells by the thymus through mechanisms that remain incompletely resolved. Here we identified the endosomal-sorting-complex-required-for-transport (ESCRT) protein CHMP5, known to be required for the formation of multivesicular bodies, as a key sensor of thresholds for signaling via the T cell antigen receptor (TCR) that was essential for T cell development. CHMP5 enabled positive selection by promoting post-selection thymocyte survival in part through stabilization of the pro-survival protein Bcl-2. Accordingly, loss of CHMP5 in thymocyte precursor cells abolished T cell development, a phenotype that was 'rescued' by genetic deletion of the pro-apoptotic protein Bim or transgenic expression of Bcl-2. Mechanistically, positive selection resulted in the stabilization of CHMP5 by inducing its interaction with the deubiquitinase USP8. Our results thus identify CHMP5 as an essential component of the post-translational machinery required for T cell development.


Assuntos
Diferenciação Celular/imunologia , Complexos Endossomais de Distribuição Requeridos para Transporte/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timócitos/imunologia , Animais , Proteína 11 Semelhante a Bcl-2/imunologia , Endopeptidases/imunologia , Immunoblotting , Imunoprecipitação , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/imunologia , Linfócitos T/citologia , Timócitos/citologia , Ubiquitina Tiolesterase/imunologia
4.
Nat Immunol ; 18(6): 665-674, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28459435

RESUMO

Tissue macrophages provide immunological defense and contribute to the establishment and maintenance of tissue homeostasis. Here we used constitutive and inducible mutagenesis to delete the nuclear transcription regulator Mecp2 in macrophages. Mice that lacked the gene encoding Mecp2, which is associated with Rett syndrome, in macrophages did not show signs of neurodevelopmental disorder but displayed spontaneous obesity, which was linked to impaired function of brown adipose tissue (BAT). Specifically, mutagenesis of a BAT-resident Cx3Cr1+ macrophage subpopulation compromised homeostatic thermogenesis but not acute, cold-induced thermogenesis. Mechanistically, malfunction of BAT in pre-obese mice with mutant macrophages was associated with diminished sympathetic innervation and local titers of norepinephrine, which resulted in lower expression of thermogenic factors by adipocytes. Mutant macrophages overexpressed the signaling receptor and ligand PlexinA4, which might contribute to the phenotype by repulsion of sympathetic axons expressing the transmembrane semaphorin Sema6A. Collectively, we report a previously unappreciated homeostatic role for macrophages in the control of tissue innervation. Disruption of this circuit in BAT resulted in metabolic imbalance.


Assuntos
Tecido Adiposo Marrom/imunologia , Macrófagos/imunologia , Proteína 2 de Ligação a Metil-CpG/genética , Sistema Nervoso Simpático/metabolismo , Termogênese/imunologia , Adipócitos Marrons , Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Animais , Axônios/metabolismo , Receptor 1 de Quimiocina CX3C , Metabolismo Energético/imunologia , Citometria de Fluxo , Homeostase , Immunoblotting , Macrófagos/metabolismo , Camundongos , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/metabolismo , Norepinefrina/metabolismo , Obesidade/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/metabolismo , Receptores de Quimiocinas/metabolismo , Semaforinas/metabolismo
5.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28553952

RESUMO

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Assuntos
Autoimunidade/genética , Infecções por Cardiovirus/genética , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon/genética , Adolescente , Adulto , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Southern Blotting , Infecções por Cardiovirus/imunologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Vírus da Encefalomiocardite/imunologia , Feminino , Predisposição Genética para Doença , Células HEK293 , Humanos , Immunoblotting , Helicase IFIH1 Induzida por Interferon/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Viroses/genética , Viroses/imunologia , Adulto Jovem
6.
Nat Immunol ; 18(9): 1025-1034, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28737753

RESUMO

Pathogenic T cells in individuals with rheumatoid arthritis (RA) infiltrate non-lymphoid tissue sites, maneuver through extracellular matrix and form lasting inflammatory microstructures. Here we found that RA T cells abundantly express the podosome scaffolding protein TKS5, which enables them to form tissue-invasive membrane structures. TKS5 overexpression was regulated by the intracellular metabolic environment of RA T cells-specifically, by reduced glycolytic flux that led to deficiencies in ATP and pyruvate. ATPlopyruvatelo conditions triggered fatty acid biosynthesis and the formation of cytoplasmic lipid droplets. Restoration of pyruvate production or inhibition of fatty acid synthesis corrected the tissue-invasiveness of RA T cells in vivo and reversed their proarthritogenic behavior. Thus, metabolic control of T cell locomotion provides new opportunities to interfere with T cell invasion into specific tissue sites.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Artrite Psoriásica/metabolismo , Artrite Reumatoide/metabolismo , Linfócitos T/metabolismo , Trifosfato de Adenosina/metabolismo , Artrite Psoriásica/imunologia , Artrite Reumatoide/imunologia , Movimento Celular/imunologia , Ácidos Graxos/biossíntese , Feminino , Perfilação da Expressão Gênica , Glicólise/imunologia , Humanos , Immunoblotting , Imuno-Histoquímica , Inflamação , Masculino , Pessoa de Meia-Idade , Ácido Pirúvico/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Membrana Sinovial/citologia , Membrana Sinovial/imunologia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Linfócitos T/imunologia
7.
Nat Immunol ; 18(9): 995-1003, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28759002

RESUMO

Among the features that distinguish type 1 innate lymphoid cells (ILC1s) from natural killer (NK) cells is a gene signature indicative of 'imprinting' by cytokines of the TGF-ß family. We studied mice in which ILC1s and NK cells lacked SMAD4, a signal transducer that facilitates the canonical signaling pathway common to all cytokines of the TGF-ß family. While SMAD4 deficiency did not affect ILC1 differentiation, NK cells unexpectedly acquired an ILC1-like gene signature and were unable to control tumor metastasis or viral infection. Mechanistically, SMAD4 restrained non-canonical TGF-ß signaling mediated by the cytokine receptor TGFßR1 in NK cells. NK cells from a SMAD4-deficient person affected by polyposis were also hyper-responsive to TGF-ß. These results identify SMAD4 as a previously unknown regulator that restricts non-canonical TGF-ß signaling in NK cells.


Assuntos
Células Matadoras Naturais/citologia , Linfopoese/genética , Proteína Smad4/genética , Fator de Crescimento Transformador beta/imunologia , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/imunologia , Animais , Estudos de Casos e Controles , Diferenciação Celular , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/imunologia , Immunoblotting , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Linfócitos/citologia , Melanoma Experimental/imunologia , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Proteína Smad4/imunologia
8.
Nat Immunol ; 18(6): 654-664, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28414311

RESUMO

In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.


Assuntos
Adipócitos Bege , Adipogenia/imunologia , Tecido Adiposo Branco/imunologia , Diferenciação Celular/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Obesidade/imunologia , Células 3T3-L1 , Adipócitos/imunologia , Adipócitos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Adesão Celular/imunologia , Dieta Hiperlipídica , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Retroalimentação , Feminino , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Integrina alfa4/genética , Macrófagos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Monócitos/imunologia , Obesidade/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Gordura Subcutânea , Linfócitos T/imunologia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Adulto Jovem
9.
Nat Immunol ; 18(7): 791-799, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28530712

RESUMO

During infection, antigen-specific T cells undergo tightly regulated developmental transitions controlled by transcriptional and post-transcriptional regulation of gene expression. We found that the microRNA miR-31 was strongly induced by activation of the T cell antigen receptor (TCR) in a pathway involving calcium and activation of the transcription factor NFAT. During chronic infection with lymphocytic choriomeningitis virus (LCMV) clone 13, miR-31-deficent mice recovered from clinical disease, while wild-type mice continued to show signs of disease. This disease phenotype was explained by the presence of larger numbers of cytokine-secreting LCMV-specific CD8+ T cells in miR-31-deficent mice than in wild-type mice. Mechanistically, miR-31 increased the sensitivity of T cells to type I interferons, which interfered with effector T cell function and increased the expression of several proteins related to T cell dysfunction during chronic infection. These studies identify miR-31 as an important regulator of T cell exhaustion in chronic infection.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , MicroRNAs/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Anticorpos Antivirais/imunologia , Infecções por Arenaviridae/genética , Linfócitos T CD8-Positivos/efeitos dos fármacos , Cálcio/metabolismo , Imunoprecipitação da Cromatina , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Immunoblotting , Interferon Tipo I/farmacologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Fatores de Transcrição NFATC/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
10.
Nat Immunol ; 18(11): 1238-1248, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28892470

RESUMO

FoxP3 conditions the transcriptional signature and functional facets of regulatory T cells (Treg cells). Its mechanism of action, whether as an activator or a repressor, has remained unclear. Here, chromatin analysis showed that FoxP3 bound active enhancer elements, not repressed chromatin, around loci over- or under-expressed in Treg cells. We evaluated the impact of a panel of FoxP3 mutants on its transcriptional activity and interactions with DNA, transcriptional cofactors and chromatin. Computational integration, confirmed by biochemical interaction and size analyses, showed that FoxP3 existed in distinct multimolecular complexes. It was active and primarily an activator when complexed with the transcriptional factors RELA, IKZF2 and KAT5. In contrast, FoxP3 was inactive when complexed with the histone methyltransferase EZH2 and transcription factors YY1 and IKZF3. The latter complex partitioned to a peripheral region of the nucleus, as shown by super-resolution microscopy. Thus, FoxP3 acts in multimodal fashion to directly activate or repress transcription, in a context- and partner-dependent manner, to govern Treg cell phenotypes.


Assuntos
Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Linfócitos T Reguladores/metabolismo , Ativação Transcricional , Animais , Células Cultivadas , DNA/genética , DNA/metabolismo , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica/métodos , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Immunoblotting , Lisina Acetiltransferase 5 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Células NIH 3T3 , Ligação Proteica , Linfócitos T Reguladores/imunologia , Transativadores/genética , Transativadores/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
11.
Nat Immunol ; 18(8): 899-910, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28604719

RESUMO

Mammalian autophagy-related 8 (Atg8) homologs consist of LC3 proteins and GABARAPs, all of which are known to be involved in canonical autophagy. In contrast, the roles of Atg8 homologs in noncanonical autophagic processes are not fully understood. Here we show a unique role of GABARAPs, in particular gamma-aminobutyric acid (GABA)-A-receptor-associated protein-like 2 (Gabarapl2; also known as Gate-16), in interferon-γ (IFN-γ)-mediated antimicrobial responses. Cells that lacked GABARAPs but not LC3 proteins and mice that lacked Gate-16 alone were defective in the IFN-γ-induced clearance of vacuolar pathogens such as Toxoplasma. Gate-16 but not LC3b specifically associated with the small GTPase ADP-ribosylation factor 1 (Arf1) to mediate uniform distribution of interferon-inducible GTPases. The lack of GABARAPs reduced Arf1 activation, which led to formation of interferon-inducible GTPase-containing aggregates and hampered recruitment of interferon-inducible GTPases to vacuolar pathogens. Thus, GABARAPs are uniquely required for antimicrobial host defense through cytosolic distribution of interferon-inducible GTPases.


Assuntos
Fator 1 de Ribosilação do ADP/imunologia , Autofagia/imunologia , Proteínas de Transporte/imunologia , Interferon gama/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Fator 1 de Ribosilação do ADP/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Família da Proteína 8 Relacionada à Autofagia , Sistemas CRISPR-Cas , Proteínas de Transporte/metabolismo , Simulação por Computador , Proteínas do Citoesqueleto/imunologia , Proteínas do Citoesqueleto/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , GTP Fosfo-Hidrolases/imunologia , GTP Fosfo-Hidrolases/metabolismo , Edição de Genes , Immunoblotting , Imunoprecipitação , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo
12.
Nat Immunol ; 18(7): 800-812, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28504697

RESUMO

An imbalance in the lineages of immunosuppressive regulatory T cells (Treg cells) and the inflammatory TH17 subset of helper T cells leads to the development of autoimmune and/or inflammatory disease. Here we found that TAZ, a coactivator of TEAD transcription factors of Hippo signaling, was expressed under TH17 cell-inducing conditions and was required for TH17 differentiation and TH17 cell-mediated inflammatory diseases. TAZ was a critical co-activator of the TH17-defining transcription factor RORγt. In addition, TAZ attenuated Treg cell development by decreasing acetylation of the Treg cell master regulator Foxp3 mediated by the histone acetyltransferase Tip60, which targeted Foxp3 for proteasomal degradation. In contrast, under Treg cell-skewing conditions, TEAD1 expression and sequestration of TAZ from the transcription factors RORγt and Foxp3 promoted Treg cell differentiation. Furthermore, deficiency in TAZ or overexpression of TEAD1 induced Treg cell differentiation, whereas expression of a transgene encoding TAZ or activation of TAZ directed TH17 cell differentiation. Our results demonstrate a pivotal role for TAZ in regulating the differentiation of Treg cells and TH17 cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Diferenciação Celular/imunologia , Colite/imunologia , Citocinas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Acetilação , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Artrite Reumatoide/imunologia , Estudos de Casos e Controles , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Células HeLa , Histona Acetiltransferases/metabolismo , Humanos , Immunoblotting , Lisina Acetiltransferase 5 , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT3/metabolismo , Síndrome de Sjogren/imunologia , Proteínas Smad/imunologia , Proteínas Smad/metabolismo , Fatores de Transcrição de Domínio TEA , Transativadores/metabolismo , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional
13.
Nat Immunol ; 18(6): 622-632, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28459433

RESUMO

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Assuntos
Calgranulina A/imunologia , Calgranulina B/imunologia , Imunidade Inata/imunologia , Monócitos/imunologia , Sepse Neonatal/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Animais Recém-Nascidos , Calgranulina A/efeitos dos fármacos , Calgranulina B/efeitos dos fármacos , Epigênese Genética , Sangue Fetal , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imunidade Inata/efeitos dos fármacos , Immunoblotting , Recém-Nascido , Inflamação , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Monócitos/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Sepse Neonatal/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor 4 Toll-Like/imunologia
14.
Nat Immunol ; 18(7): 733-743, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28481329

RESUMO

The transcription regulator YAP controls organ size by regulating cell growth, proliferation and apoptosis. However, whether YAP has a role in innate antiviral immunity is largely unknown. Here we found that YAP negatively regulated an antiviral immune response. YAP deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in vivo. YAP blocked dimerization of the transcription factor IRF3 and impeded translocation of IRF3 to the nucleus after viral infection. Notably, virus-activated kinase IKKɛ phosphorylated YAP at Ser403 and thereby triggered degradation of YAP in lysosomes and, consequently, relief of YAP-mediated inhibition of the cellular antiviral response. These findings not only establish YAP as a modulator of the activation of IRF3 but also identify a previously unknown regulatory mechanism independent of the kinases Hippo and LATS via which YAP is controlled by the innate immune pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Fibroblastos/imunologia , Quinase I-kappa B/metabolismo , Imunidade Inata/imunologia , Lisossomos/metabolismo , Macrófagos/imunologia , Fosfoproteínas/imunologia , Infecções por Rhabdoviridae/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Imunofluorescência , Edição de Genes , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , Interferon beta/imunologia , Pulmão/imunologia , Pulmão/patologia , Camundongos , Microscopia Confocal , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções por Rhabdoviridae/patologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia , Vesiculovirus , Carga Viral , Proteínas de Sinalização YAP
15.
Nat Immunol ; 17(3): 304-14, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26829766

RESUMO

The role of anergy, an acquired state of T cell functional unresponsiveness, in natural peripheral tolerance remains unclear. In this study, we found that anergy was selectively induced in fetal antigen-specific maternal CD4(+) T cells during pregnancy. A naturally occurring subpopulation of anergic polyclonal CD4(+) T cells, enriched for self antigen-specific T cell antigen receptors, was also present in healthy hosts. Neuropilin-1 expression in anergic conventional CD4(+) T cells was associated with hypomethylation of genes related to thymic regulatory T cells (Treg cells), and this correlated with their ability to differentiate into Foxp3(+) Treg cells that suppressed immunopathology. Thus, our data suggest that not only is anergy induction important in preventing autoimmunity but also it generates the precursors for peripheral Treg cell differentiation.


Assuntos
Autoimunidade/imunologia , Diferenciação Celular/imunologia , Anergia Clonal/imunologia , Histocompatibilidade Materno-Fetal/imunologia , Tolerância Periférica/imunologia , Células Precursoras de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Artrite Experimental/imunologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Immunoblotting , Masculino , Camundongos , Camundongos Knockout , Neuropilina-1/metabolismo , Gravidez , Receptores de Antígenos de Linfócitos T/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tolerância a Antígenos Próprios , Timócitos/imunologia
16.
Nat Immunol ; 17(3): 286-96, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26829767

RESUMO

SHARPIN forms a linear-ubiquitin-chain-assembly complex that promotes signaling via the transcription factor NF-κB. SHARPIN deficiency leads to progressive multi-organ inflammation and immune system malfunction, but how SHARPIN regulates T cell responses is unclear. Here we found that SHARPIN deficiency resulted in a substantial reduction in the number of and defective function of regulatory T cells (Treg cells). Transfer of SHARPIN-sufficient Treg cells into SHARPIN-deficient mice considerably alleviated their systemic inflammation. SHARPIN-deficient T cells displayed enhanced proximal signaling via the T cell antigen receptor (TCR) without an effect on the activation of NF-κB. SHARPIN conjugated with Lys63 (K63)-linked ubiquitin chains, which led to inhibition of the association of TCRζ with the signaling kinase Zap70; this affected the generation of Treg cells. Our study therefore identifies a role for SHARPIN in TCR signaling whereby it maintains immunological homeostasis and tolerance by regulating Treg cells.


Assuntos
Proteínas de Transporte/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proteínas de Transporte/genética , Colite/imunologia , Citocinas/imunologia , Feminino , Citometria de Fluxo , Humanos , Tolerância Imunológica/imunologia , Immunoblotting , Imunoprecipitação , Técnicas In Vitro , Inflamação , Peptídeos e Proteínas de Sinalização Intracelular , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/imunologia , Transdução de Sinais , Ubiquitinação , Proteína-Tirosina Quinase ZAP-70/imunologia
17.
Nat Immunol ; 17(4): 379-86, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26901151

RESUMO

The T cell antigen receptor (TCR) is unique in that its affinity for ligand is unknown before encounter and can vary by orders of magnitude. How the immune system regulates individual T cells that display very different reactivity to antigen remains unclear. Here we found that activated CD4(+) T cells, at the peak of clonal expansion, persistently downregulated their TCR expression in proportion to the strength of the initial antigen recognition. This programmed response increased the threshold for cytokine production and recall proliferation in a clone-specific manner and ultimately excluded clones with the highest antigen reactivity. Thus, programmed downregulation of TCR expression represents a negative feedback mechanism for constraining T cell effector function with a suitable time delay to thereby allow pathogen control while avoiding excess inflammatory damage.


Assuntos
Regulação para Baixo , Listeriose/imunologia , Receptores de Antígenos de Linfócitos T/genética , Células Th1/imunologia , Tuberculose Pulmonar/imunologia , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Immunoblotting , Listeria monocytogenes , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Mycobacterium tuberculosis , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T , Transcriptoma
18.
Nat Immunol ; 17(1): 95-103, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26523864

RESUMO

Aerobic glycolysis regulates T cell function. However, whether and how primary cancer alters T cell glycolytic metabolism and affects tumor immunity in cancer patients remains a question. Here we found that ovarian cancers imposed glucose restriction on T cells and dampened their function via maintaining high expression of microRNAs miR-101 and miR-26a, which constrained expression of the methyltransferase EZH2. EZH2 activated the Notch pathway by suppressing Notch repressors Numb and Fbxw7 via trimethylation of histone H3 at Lys27 and, consequently, stimulated T cell polyfunctional cytokine expression and promoted their survival via Bcl-2 signaling. Moreover, small hairpin RNA-mediated knockdown of human EZH2 in T cells elicited poor antitumor immunity. EZH2(+)CD8(+) T cells were associated with improved survival in patients. Together, these data unveil a metabolic target and mechanism of cancer immune evasion.


Assuntos
Regulação Neoplásica da Expressão Gênica/imunologia , MicroRNAs , Neoplasias/imunologia , Complexo Repressor Polycomb 2/imunologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Animais , Separação Celular , Imunoprecipitação da Cromatina , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Citometria de Fluxo , Imunofluorescência , Glicólise , Humanos , Immunoblotting , Melanoma Experimental/imunologia , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Análise Serial de Tecidos , Transfecção
19.
Nat Immunol ; 17(1): 104-12, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26551880

RESUMO

We used high-resolution mass spectrometry to map the cytotoxic T lymphocyte (CTL) proteome and the effect of the metabolic checkpoint kinase mTORC1 on CTLs. The CTL proteome was dominated by metabolic regulators and granzymes, and mTORC1 selectively repressed and promoted expression of a subset of CTL proteins (~10%). These included key CTL effector molecules, signaling proteins and a subset of metabolic enzymes. Proteomic data highlighted the potential for negative control of the production of phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) by mTORC1 in CTLs. mTORC1 repressed PtdIns(3,4,5)P3 production and determined the requirement for mTORC2 in activation of the kinase Akt. Our unbiased proteomic analysis thus provides comprehensive understanding of CTL identity and the control of CTL function by mTORC1.


Assuntos
Complexos Multiproteicos/metabolismo , Proteoma/imunologia , Linfócitos T Citotóxicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Células Cultivadas , Cromatografia , Ensaio de Imunoadsorção Enzimática , Feminino , Immunoblotting , Masculino , Espectrometria de Massas , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Linfócitos T Citotóxicos/imunologia , Serina-Treonina Quinases TOR/imunologia
20.
Nat Immunol ; 17(5): 531-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27043408

RESUMO

The antigen-presenting molecule MR1 presents vitamin B-related antigens (VitB antigens) to mucosal-associated invariant T (MAIT) cells through an uncharacterized pathway. We show that MR1, unlike other antigen-presenting molecules, does not constitutively present self-ligands. In the steady state it accumulates in a ligand-receptive conformation within the endoplasmic reticulum. VitB antigens reach this location and form a Schiff base with MR1, triggering a 'molecular switch' that allows MR1-VitB antigen complexes to traffic to the plasma membrane. These complexes are endocytosed with kinetics independent of the affinity of the MR1-ligand interaction and are degraded intracellularly, although some MR1 molecules acquire new ligands during passage through endosomes and recycle back to the surface. MR1 antigen presentation is characterized by a rapid 'off-on-off' mechanism that is strictly dependent on antigen availability.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Transdução de Sinais/imunologia , Antígenos/metabolismo , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Endocitose/imunologia , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Endossomos/imunologia , Endossomos/metabolismo , Células HeLa , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Immunoblotting , Espaço Intracelular/imunologia , Espaço Intracelular/metabolismo , Microscopia Confocal , Antígenos de Histocompatibilidade Menor , Ligação Proteica/imunologia , Transporte Proteico/imunologia , Complexo Vitamínico B/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA