Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45.493
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 18(9): 1035-1045, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28759003

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , Chromatin/metabolism , Forkhead Transcription Factors/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , T-Lymphocytes, Regulatory , Animals , CRISPR-Cas Systems , Cytokines/immunology , Flow Cytometry , Gene Expression Regulation , Immunoblotting , In Vitro Techniques , Methylation , Mice
2.
Nat Immunol ; 18(7): 780-790, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553951

ABSTRACT

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.


Subject(s)
Cell Differentiation/immunology , Endosomal Sorting Complexes Required for Transport/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , Bcl-2-Like Protein 11/immunology , Endopeptidases/immunology , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/immunology , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymocytes/cytology , Ubiquitin Thiolesterase/immunology
3.
Nat Immunol ; 18(11): 1197-1206, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28920951

ABSTRACT

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.


Subject(s)
Glucose/metabolism , Glycolysis , Killer Cells, Natural/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Cell Proliferation , Cytokines/metabolism , Flow Cytometry , Humans , Immunoblotting , Killer Cells, Natural/immunology , Lipids/biosynthesis , Oxidative Phosphorylation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
4.
Nat Immunol ; 18(9): 1025-1034, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28737753

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Arthritis, Psoriatic/metabolism , Arthritis, Rheumatoid/metabolism , T-Lymphocytes/metabolism , Adenosine Triphosphate/metabolism , Arthritis, Psoriatic/immunology , Arthritis, Rheumatoid/immunology , Cell Movement/immunology , Fatty Acids/biosynthesis , Female , Gene Expression Profiling , Glycolysis/immunology , Humans , Immunoblotting , Immunohistochemistry , Inflammation , Male , Middle Aged , Pyruvic Acid/metabolism , Real-Time Polymerase Chain Reaction , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , T-Lymphocytes/immunology
5.
Nat Immunol ; 18(9): 995-1003, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28759002

ABSTRACT

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.


Subject(s)
Killer Cells, Natural/cytology , Lymphopoiesis/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta/immunology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/immunology , Animals , Case-Control Studies , Cell Differentiation , Gene Expression Profiling , Humans , Immunity, Innate/immunology , Immunoblotting , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocytes/cytology , Melanoma, Experimental/immunology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Smad4 Protein/immunology
6.
Nat Immunol ; 18(7): 791-799, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530712

ABSTRACT

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.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , MicroRNAs/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Viral/immunology , Arenaviridae Infections/genetics , CD8-Positive T-Lymphocytes/drug effects , Calcium/metabolism , Chromatin Immunoprecipitation , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Immunoblotting , Interferon Type I/pharmacology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , MicroRNAs/genetics , NFATC Transcription Factors/metabolism , Real-Time Polymerase Chain Reaction
7.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553952

ABSTRACT

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.


Subject(s)
Autoimmunity/genetics , Cardiovirus Infections/genetics , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/genetics , Adolescent , Adult , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/immunology , Blotting, Southern , Cardiovirus Infections/immunology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Encephalomyocarditis virus/immunology , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Immunoblotting , Interferon-Induced Helicase, IFIH1/immunology , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Diseases/genetics , Virus Diseases/immunology , Young Adult
8.
Nat Immunol ; 18(6): 665-674, 2017 06.
Article in English | MEDLINE | ID: mdl-28459435

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown/immunology , Macrophages/immunology , Methyl-CpG-Binding Protein 2/genetics , Sympathetic Nervous System/metabolism , Thermogenesis/immunology , Adipocytes, Brown , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Animals , Axons/metabolism , CX3C Chemokine Receptor 1 , Energy Metabolism/immunology , Flow Cytometry , Homeostasis , Immunoblotting , Macrophages/metabolism , Mice , Mutagenesis, Site-Directed , Nerve Tissue Proteins/metabolism , Norepinephrine/metabolism , Obesity/genetics , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/metabolism , Receptors, Chemokine/metabolism , Semaphorins/metabolism
9.
Nat Immunol ; 18(8): 899-910, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28604719

ABSTRACT

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.


Subject(s)
ADP-Ribosylation Factor 1/immunology , Autophagy/immunology , Carrier Proteins/immunology , Interferon-gamma/immunology , Microtubule-Associated Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , ADP-Ribosylation Factor 1/metabolism , Animals , Apoptosis Regulatory Proteins , Autophagy-Related Protein 8 Family , CRISPR-Cas Systems , Carrier Proteins/metabolism , Computer Simulation , Cytoskeletal Proteins/immunology , Cytoskeletal Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , GTP Phosphohydrolases/immunology , GTP Phosphohydrolases/metabolism , Gene Editing , Immunoblotting , Immunoprecipitation , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism
10.
Nat Immunol ; 18(11): 1238-1248, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28892470

ABSTRACT

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.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation , T-Lymphocytes, Regulatory/metabolism , Transcriptional Activation , Animals , Cells, Cultured , DNA/genetics , DNA/metabolism , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling/methods , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Immunoblotting , Lysine Acetyltransferase 5 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , NIH 3T3 Cells , Protein Binding , T-Lymphocytes, Regulatory/immunology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
11.
Nat Immunol ; 18(6): 654-664, 2017 06.
Article in English | MEDLINE | ID: mdl-28414311

ABSTRACT

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.


Subject(s)
Adipocytes, Beige , Adipogenesis/immunology , Adipose Tissue, White/immunology , Cell Differentiation/immunology , Inflammation/immunology , Macrophages/immunology , Obesity/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Adhesion/immunology , Diet, High-Fat , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback , Female , Gene Knockdown Techniques , Humans , Immunoblotting , Integrin alpha4/genetics , Macrophages/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Obesity/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Subcutaneous Fat , T-Lymphocytes/immunology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Young Adult
12.
Nat Immunol ; 18(7): 733-743, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28481329

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Fibroblasts/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/immunology , Lysosomes/metabolism , Macrophages/immunology , Phosphoproteins/immunology , Rhabdoviridae Infections/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CRISPR-Cas Systems , Cell Cycle Proteins , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Fluorescent Antibody Technique , Gene Editing , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Lung/immunology , Lung/pathology , Mice , Microscopy, Confocal , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdoviridae Infections/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Vesiculovirus , Viral Load , YAP-Signaling Proteins
13.
Nat Immunol ; 18(6): 622-632, 2017 06.
Article in English | MEDLINE | ID: mdl-28459433

ABSTRACT

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.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Immunity, Innate/immunology , Monocytes/immunology , Neonatal Sepsis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Calgranulin A/drug effects , Calgranulin B/drug effects , Epigenesis, Genetic , Fetal Blood , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Immunoblotting , Infant, Newborn , Inflammation , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Monocytes/drug effects , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neonatal Sepsis/genetics , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/immunology
14.
Nat Immunol ; 18(7): 800-812, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28504697

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Differentiation/immunology , Colitis/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intracellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Arthritis, Rheumatoid/immunology , Case-Control Studies , Chromatin Immunoprecipitation , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Immunoblotting , Lysine Acetyltransferase 5 , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sjogren's Syndrome/immunology , Smad Proteins/immunology , Smad Proteins/metabolism , TEA Domain Transcription Factors , Trans-Activators/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins
15.
Nat Immunol ; 17(1): 104-12, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26551880

ABSTRACT

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.


Subject(s)
Multiprotein Complexes/metabolism , Proteome/immunology , T-Lymphocytes, Cytotoxic/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Chromatography , Enzyme-Linked Immunosorbent Assay , Female , Immunoblotting , Male , Mass Spectrometry , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/immunology , Oligonucleotide Array Sequence Analysis , T-Lymphocytes, Cytotoxic/immunology , TOR Serine-Threonine Kinases/immunology
16.
Nat Immunol ; 17(1): 95-103, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26523864

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , MicroRNAs , Neoplasms/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Animals , Cell Separation , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycolysis , Humans , Immunoblotting , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Transfection
17.
Nat Immunol ; 17(4): 379-86, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901151

ABSTRACT

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.


Subject(s)
Down-Regulation , Listeriosis/immunology , Receptors, Antigen, T-Cell/genetics , Th1 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Immunoblotting , Listeria monocytogenes , Lymphocyte Activation , Mice , Mice, Transgenic , Mycobacterium tuberculosis , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes , Transcriptome
18.
Nat Immunol ; 17(3): 304-14, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829766

ABSTRACT

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.


Subject(s)
Autoimmunity/immunology , Cell Differentiation/immunology , Clonal Anergy/immunology , Histocompatibility, Maternal-Fetal/immunology , Peripheral Tolerance/immunology , Precursor Cells, T-Lymphoid/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Arthritis, Experimental/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Forkhead Transcription Factors/immunology , Genes, T-Cell Receptor alpha , Immunoblotting , Male , Mice , Mice, Knockout , Neuropilin-1/metabolism , Pregnancy , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance , Thymocytes/immunology
19.
Nat Immunol ; 17(3): 286-96, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829767

ABSTRACT

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.


Subject(s)
Carrier Proteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Colitis/immunology , Cytokines/immunology , Female , Flow Cytometry , Humans , Immune Tolerance/immunology , Immunoblotting , Immunoprecipitation , In Vitro Techniques , Inflammation , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/immunology , Signal Transduction , Ubiquitination , ZAP-70 Protein-Tyrosine Kinase/immunology
20.
Nat Immunol ; 17(5): 523-30, 2016 May.
Article in English | MEDLINE | ID: mdl-26998762

ABSTRACT

14-3-3 proteins regulate biological processes by binding to phosphorylated serine or phosphorylated threonine motifs of cellular proteins. Among the 14-3-3 proteins, 14-3-3ɛ serves a crucial function in antiviral immunity by mediating the cytosol-to-mitochondrial membrane translocation of the pathogen sensor RIG-I. Here we found that the NS3 protein of dengue virus (DV) bound to 14-3-3ɛ and prevented translocation of RIG-I to the adaptor MAVS and thereby blocked antiviral signaling. Intriguingly, a highly conserved phosphomimetic RxEP motif in NS3 was essential for the binding of 14-3-3ɛ. A recombinant mutant DV deficient in binding to 14-3-3ɛ showed impairment in antagonism of RIG-I and elicited a markedly augmented innate immune response and enhanced T cell activation. Our work reveals a novel phosphomimetic-based mechanism for viral antagonism of 14-3-3-mediated immunity, which might guide the rational design of therapeutics.


Subject(s)
14-3-3 Proteins/immunology , DEAD-box RNA Helicases/immunology , Immunity, Innate/immunology , Serine Endopeptidases/immunology , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , HEK293 Cells , Humans , Immunoblotting , Microscopy, Confocal , Phosphorylation/immunology , RNA Interference/immunology , Receptors, Immunologic , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL