Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Nat Commun ; 15(1): 899, 2024 Feb 06.
Article En | MEDLINE | ID: mdl-38321013

Antigen-specific regulatory T cells (Tregs) suppress pathogenic autoreactivity and are potential therapeutic candidates for autoimmune diseases such as systemic lupus erythematosus (SLE). Lupus nephritis is associated with autoreactivity to the Smith (Sm) autoantigen and the human leucocyte antigen (HLA)-DR15 haplotype; hence, we investigated the potential of Sm-specific Tregs (Sm-Tregs) to suppress disease. Here we identify a HLA-DR15 restricted immunodominant Sm T cell epitope using biophysical affinity binding assays, then identify high-affinity Sm-specific T cell receptors (TCRs) using high-throughput single-cell sequencing. Using lentiviral vectors, we transduce our lead Sm-specific TCR into Tregs derived from patients with SLE who are anti-Sm and HLA-DR15 positive. Compared with polyclonal mock-transduced Tregs, Sm-Tregs potently suppress Sm-specific pro-inflammatory responses in vitro and suppress disease progression in a humanized mouse model of lupus nephritis. These results show that Sm-Tregs are a promising therapy for SLE.


Lupus Erythematosus, Systemic , Lupus Nephritis , Mice , Animals , Humans , T-Lymphocytes, Regulatory , Autoantigens/metabolism
2.
Front Immunol ; 14: 1235222, 2023.
Article En | MEDLINE | ID: mdl-37965348

Conventional type 1 dendritic cells (DC1) contribute to the development of pathogenic T helper type 1 (Th1) cells in part via the production of the proinflammatory cytokine interleukin-12. Thus, depletion of DC1 has the potential to dampen autoimmune responses. Here, we developed X-C motif chemokine receptor 1 (XCR1)-specific chimeric antigen receptor (CAR)-T cells and CAR-Tregs that specifically targeted DC1. XCR1 CAR-T cells were successfully generated as CD4+ and CD8+ T cells, expressed XCR1 CAR efficiently, and induced XCR1-dependent activation, cytokine production and proliferation. XCR1 CAR-T cells selectively depleted DC1 when transferred into RAG2-/- mice with a compensatory increase in conventional type 2 DC (DC2) and plasmacytoid DC (pDC). XCR1 CAR-T cell-mediated depletion of DC1 modestly suppressed the onset of Th1-driven experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Diphtheria toxin-mediated DC1 depletion in XCR1-diphtheria toxin receptor mice also suppressed EAE, suggesting that DC1 depletion was responsible for EAE suppression. XCR1 CAR-Tregs were successfully generated and suppressed effector T cells in the presence of XCR1+ cells. Therapeutic treatment with XCR1 CAR-Tregs suppressed Th1-driven EAE. Therefore, we conclude that depletion of DC1 with XCR1 CAR-T cells or immune suppression with XCR1 CAR-Tregs can modestly suppress Th1-driven EAE.


Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , CD8-Positive T-Lymphocytes/pathology , Cytokines/metabolism , Th1 Cells , Dendritic Cells
3.
Front Immunol ; 14: 1257652, 2023.
Article En | MEDLINE | ID: mdl-37809101

Interleukin-2 (IL-2), along with T-cell receptor (TCR) signaling, are required to control regulatory T cell (Treg) homeostasis and function in vivo. Due to the heightened sensitivity to IL-2, Tregs retain the ability to respond to low-dose or attenuated forms of IL-2, as currently being developed for clinical use to treat inflammatory diseases. While attenuated IL-2 increases Treg selectivity, the question remains as to whether a weakened IL-2 signal sufficiently enhances Treg suppressive function(s) toward disease modification. To understand this question, we characterized the in vivo activity and transcriptomic profiles of two different attenuated IL-2 muteins in comparison with wildtype (WT) IL-2. Our study showed that, in addition to favoring Tregs, the attenuated muteins induced disproportionately robust effects on Treg activation and conversion to effector Treg (eTreg) phenotype. Our data furthermore suggested that Tregs activated by attenuated IL-2 muteins showed reduced dependence on TCR signal, at least in part due to the enhanced ability of IL-2 muteins to amplify the TCR signal in vivo. These results point to a new paradigm wherein IL-2 influences Tregs' sensitivity to antigenic signal, and that the combination effect may be leveraged for therapeutic use of attenuated IL-2 muteins.


Interleukin-2 , Receptors, Antigen, T-Cell , T-Lymphocytes, Regulatory , Homeostasis , Interleukin-2/genetics , Interleukin-2/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Humans
4.
Front Immunol ; 11: 1106, 2020.
Article En | MEDLINE | ID: mdl-32582190

Among all T and NK cell subsets, regulatory T (Treg) cells typically respond to the lowest concentrations of IL-2 due to elevated surface expression of the IL-2R alpha chain (IL2RA; CD25) and the high affinity IL-2 receptor (IL-2R) complex. This enhanced sensitivity forms the basis for low-dose (LD) IL-2 therapy for the treatment of inflammatory diseases, where efficacy correlates with increased Treg cell number and expression of functional markers. Despite strong preclinical support for this approach, moderate and variable clinical efficacy has raised concerns that adequate Treg selectivity still cannot be achieved with LD IL-2, and/or that doses are too low to stimulate effective Treg-mediated suppression within tissues. This has prompted development of IL-2 variants with greater Treg selectivity, achieved through attenuated affinity for the signaling chains of the IL-2R complex (IL2RB or CD122 and IL2RG or CD132) and, consequently, greater reliance on high CD25 levels for full receptor binding and signaling. While certain IL-2 variants have advanced to the clinic, it remains unknown if the full range of IL-2R signaling potency and Treg-selectivity observed with low concentrations of wildtype IL-2 can be sufficiently recapitulated with attenuated IL-2 muteins at high concentrations. Using a panel of engineered IL-2 muteins, we investigated how a range of IL-2R signaling intensity, benchmarked by the degree of STAT5 phosphorylation, relates to biologically relevant Treg cell responses such as proliferation, lineage and phenotypic marker expression, and suppressor function. Our results demonstrate that a surprisingly wide dynamic range of IL-2R signaling intensity leads to productive biological responses in Treg cells, with negligible STAT5 phosphorylation associating with nearly complete downstream effects such as Treg proliferation and suppressor activity. Furthermore, we show with both in vitro and humanized mouse in vivo systems that different biological responses in Treg cells require different minimal IL-2R signaling thresholds. Our findings suggest that more than minimal IL-2R signaling, beyond that capable of driving Treg cell proliferation, may be required to fully enhance Treg cell stability and suppressor function in vivo.


Interleukin-2/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Interleukin-2/metabolism , Mice , Protein Binding , Protein Engineering , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Structure-Activity Relationship
5.
J Leukoc Biol ; 107(6): 917-932, 2020 06.
Article En | MEDLINE | ID: mdl-32272497

T cells are crucial for the success of immune-based cancer therapy. Reinvigorating antitumor T cell activity by blocking checkpoint inhibitory receptors has provided clinical benefits for many cancer patients. However, the efficacy of these treatments varies in cancer patients and the mechanisms underlying these diverse responses remain elusive. The density and status of tumor-infiltrating T cells have been shown to positively correlate with patient response to checkpoint blockades. Therefore, further understanding of the heterogeneity, clonal expansion, migration, and effector functions of tumor-infiltrating T cells will provide fundamental insights into antitumor immune responses. To this end, recent advances in single-cell RNA sequencing technology have enabled profound and extensive characterization of intratumoral immune cells and have improved our understanding of their dynamic relationships. Here, we summarize recent progress in single-cell RNA sequencing technology and current strategies to uncover heterogeneous tumor-infiltrating T cell subsets. In particular, we discuss how the coupling of deep transcriptome information with T cell receptor (TCR)-based lineage tracing has furthered our understanding of intratumoral T cell populations. We also discuss the functional implications of various T cell subsets in tumors and highlight the identification of novel T cell markers with therapeutic or prognostic potential.


Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , RNA, Neoplasm/genetics , T-Lymphocyte Subsets/immunology , Transcriptome , Cell Communication/genetics , Cell Communication/immunology , Gene Expression , Genetic Heterogeneity , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasms/genetics , Neoplasms/pathology , RNA, Neoplasm/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sequence Analysis, RNA , Single-Cell Analysis , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/pathology , Tumor Microenvironment/immunology
6.
Cell Rep ; 27(5): 1472-1486.e5, 2019 04 30.
Article En | MEDLINE | ID: mdl-31042474

The immunoglobulin heavy chain (Igh) locus features a dynamic chromatin landscape to promote class switch recombination (CSR), yet the mechanisms that regulate this landscape remain poorly understood. CHD4, a component of the chromatin remodeling NuRD complex, directly binds H3K9me3, an epigenetic mark present at the Igh locus during CSR. We find that CHD4 is essential for early B cell development but is dispensable for the homeostatic maintenance of mature, naive B cells. However, loss of CHD4 in mature B cells impairs CSR because of suboptimal targeting of AID to the Igh locus. Additionally, we find that CHD4 represses p53 expression to promote B cell proliferation. This work reveals distinct roles for CHD4 in B cell development and CSR and links the H3K9me3 epigenetic mark with AID recruitment to the Igh locus.


B-Lymphocytes/immunology , Cell Proliferation , DNA Helicases/genetics , Immunoglobulin Class Switching , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation , Cells, Cultured , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Genes, Immunoglobulin Heavy Chain , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
PLoS One ; 13(2): e0191837, 2018.
Article En | MEDLINE | ID: mdl-29401462

Mucosal-associated invariant T (MAIT) cells are an abundant class of innate T cells restricted by the MHC I-related molecule MR1. MAIT cells can recognize bacterially-derived metabolic intermediates from the riboflavin pathway presented by MR1 and are postulated to play a role in innate antibacterial immunity through production of cytokines and direct bacterial killing. MR1 tetramers, typically stabilized by the adduct of 5-amino-6-D-ribitylaminouracil (5-A-RU) and methylglyoxal (MeG), are important tools for the study of MAIT cells. A long-standing problem with 5-A-RU is that it is unstable upon storage. Herein we report an efficient synthetic approach to the HCl salt of this ligand, which has improved stability during storage. We also show that synthetic 5-A-RU•HCl produced by this method may be used in protocols for the stimulation of human MAIT cells and production of both human and mouse MR1 tetramers for MAIT cell identification.


Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens/metabolism , Ribitol/analogs & derivatives , Uracil/analogs & derivatives , Humans , Immunity, Innate , Ligands , Mucosal-Associated Invariant T Cells/immunology , Ribitol/chemical synthesis , Ribitol/metabolism , Uracil/chemical synthesis , Uracil/metabolism
9.
Proc Natl Acad Sci U S A ; 114(31): 8354-8359, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28724724

DNA double-strand breaks (DSBs) serve as obligatory intermediates for Ig heavy chain (Igh) class switch recombination (CSR). The mechanisms by which DSBs are resolved to promote long-range DNA end-joining while suppressing genomic instability inherently associated with DSBs are yet to be fully elucidated. Here, we use a targeted short-hairpin RNA screen in a B-cell lymphoma line to identify the BRCT-domain protein BRIT1 as an effector of CSR. We show that conditional genetic deletion of BRIT1 in mice leads to a marked increase in unrepaired Igh breaks and a significant reduction in CSR in ex vivo activated splenic B cells. We find that the C-terminal tandem BRCT domains of BRIT1 facilitate its interaction with phosphorylated H2AX and that BRIT1 is recruited to the Igh locus in an activation-induced cytidine deaminase (AID) and H2AX-dependent fashion. Finally, we demonstrate that depletion of another BRCT-domain protein, MDC1, in BRIT1-deleted B cells increases the severity of CSR defect over what is observed upon loss of either protein alone. Our results identify BRIT1 as a factor in CSR and demonstrate that multiple BRCT-domain proteins contribute to optimal resolution of AID-induced DSBs.

10.
Nature ; 546(7658): 421-425, 2017 06 15.
Article En | MEDLINE | ID: mdl-28607488

Adaptive immune responses are tailored to different types of pathogens through differentiation of naive CD4 T cells into functionally distinct subsets of effector T cells (T helper 1 (TH1), TH2, and TH17) defined by expression of the key transcription factors T-bet, GATA3, and RORγt, respectively. Regulatory T (Treg) cells comprise a distinct anti-inflammatory lineage specified by the X-linked transcription factor Foxp3 (refs 2, 3). Paradoxically, some activated Treg cells express the aforementioned effector CD4 T cell transcription factors, which have been suggested to provide Treg cells with enhanced suppressive capacity. Whether expression of these factors in Treg cells-as in effector T cells-is indicative of heterogeneity of functionally discrete and stable differentiation states, or conversely may be readily reversible, is unknown. Here we demonstrate that expression of the TH1-associated transcription factor T-bet in mouse Treg cells, induced at steady state and following infection, gradually becomes highly stable even under non-permissive conditions. Loss of function or elimination of T-bet-expressing Treg cells-but not of T-bet expression in Treg cells-resulted in severe TH1 autoimmunity. Conversely, following depletion of T-bet- Treg cells, the remaining T-bet+ cells specifically inhibited TH1 and CD8 T cell activation consistent with their co-localization with T-bet+ effector T cells. These results suggest that T-bet+ Treg cells have an essential immunosuppressive function and indicate that Treg cell functional heterogeneity is a critical feature of immunological tolerance.


Immune Tolerance/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Female , Lymphocyte Activation , Male , Mice , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
11.
J Immunol ; 198(5): 1823-1837, 2017 03 01.
Article En | MEDLINE | ID: mdl-28100678

T cell response magnitudes increase with increasing antigenic dosage. However, it is unclear whether ligand density only modulates the proportions of responding ligand-specific T cells or also alters responses at the single cell level. Using brief (3 h) exposure of TCR-transgenic mouse CD8 T cells in vitro to varying densities of cognate peptide-MHC ligand followed by ligand-free culture in IL-2, we found that ligand density determined the frequencies of responding cells but not the expression levels of the early activation marker molecule, CD69. Cells with low glucose uptake capacity and low protein synthesis rates were less ligand-sensitive, implicating metabolic competence in the response heterogeneity of CD8 T cell populations. Although most responding cells proliferated, ligand density was associated with time of entry into proliferation and with the extent of cell surface TCR downmodulation. TCR internalization was associated, regardless of the ligand density, with rapidity of c-myc induction, loss of the cell cycle inhibitor p27kip1, metabolic reprogramming, and cell cycle entry. A low affinity peptide ligand behaved, regardless of ligand density, like a low density, high affinity ligand in all these parameters. Inhibition of signaling after ligand exposure selectively delayed proliferation in cells with internalized TCRs. Finally, internalized TCRs continued to signal and genetic modification of TCR internalization and trafficking altered the duration of signaling in a T cell hybridoma. Together, our findings indicate that heterogeneity among responding CD8 T cell populations in their ability to respond to TCR-mediated stimulation and internalize TCRs mediates detection of ligand density or affinity, contributing to graded response magnitudes.


CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line , Dendritic Cells/immunology , Glucose/metabolism , Interleukin-2/pharmacology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Ligands , Lymphocyte Activation/immunology , Mice , Peptides/metabolism , Peptides/pharmacology , Receptors, Antigen, T-Cell/immunology
12.
J Exp Med ; 214(1): 197-208, 2017 01.
Article En | MEDLINE | ID: mdl-28011866

Generation of cellular heterogeneity is an essential feature of the adaptive immune system. This is best exemplified during humoral immune response when an expanding B cell clone assumes multiple cell fates, including class-switched B cells, antibody-secreting plasma cells, and memory B cells. Although each cell type is essential for immunity, their generation must be exquisitely controlled because a class-switched B cell cannot revert back to the parent isotype, and a terminally differentiated plasma cell cannot contribute to the memory pool. In this study, we show that an environmental sensor, the aryl hydrocarbon receptor (AhR) is highly induced upon B cell activation and serves a critical role in regulating activation-induced cell fate outcomes. We find that AhR negatively regulates class-switch recombination ex vivo by altering activation-induced cytidine deaminase expression. We further demonstrate that AhR suppresses class switching in vivo after influenza virus infection and immunization with model antigens. In addition, by regulating Blimp-1 expression via Bach2, AhR represses differentiation of B cells into plasmablasts ex vivo and antibody-secreting plasma cells in vivo. These experiments suggest that AhR serves as a molecular rheostat in B cells to brake the effector response, possibly to facilitate optimal recall responses. Thus, AhR might represent a novel molecular target for manipulation of B cell responses during vaccination.


B-Lymphocytes/physiology , Receptors, Aryl Hydrocarbon/physiology , Animals , Cell Differentiation , Cytidine Deaminase/physiology , Female , Immunoglobulin Class Switching , Influenza A Virus, H1N1 Subtype/immunology , Male , Mice , Mice, Inbred C57BL , Plasma Cells/cytology , Polychlorinated Dibenzodioxins/pharmacology , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes/physiology , Transcription Factors/physiology
13.
Immunity ; 43(1): 52-64, 2015 Jul 21.
Article En | MEDLINE | ID: mdl-26163372

MicroRNA (miRNA)-dependent regulation of gene expression confers robustness to cellular phenotypes and controls responses to extracellular stimuli. Although a single miRNA can regulate expression of hundreds of target genes, it is unclear whether any of its distinct biological functions can be due to the regulation of a single target. To explore in vivo the function of a single miRNA-mRNA interaction, we mutated the 3' UTR of a major miR-155 target (SOCS1) to specifically disrupt its regulation by miR-155. We found that under physiologic conditions and during autoimmune inflammation or viral infection, some immunological functions of miR-155 were fully or largely attributable to the regulation of SOCS1, whereas others could be accounted only partially or not at all by this interaction. Our data suggest that the role of a single miRNA-mRNA interaction is dependent on cell type and biological context.


CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , MicroRNAs/genetics , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes, Regulatory/immunology , 3' Untranslated Regions/genetics , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Profiling , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Killer Cells, Natural/transplantation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Mutation , RNA, Messenger/genetics , Suppressor of Cytokine Signaling 1 Protein
14.
J Clin Invest ; 123(3): 939-44, 2013 Mar.
Article En | MEDLINE | ID: mdl-23454755

Tregs have been implicated in control of homeostasis in the immune system and beyond. These cells restrain inflammatory responses to self antigens, commensal microorganisms, allergens, and pathogens and adapt their homeostatic and functional capabilities to a particular environment. In this review, we discuss a general model of integration of environmental cues by Tregs in which specialized Treg homeostatic, migratory, and suppression programs are established in dynamically changing inflammatory environments by maintaining an optimal threshold of activation of transcription factors involved in regulation of the corresponding type of effector immune responses.


Inflammation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cytokines/metabolism , Cytokines/physiology , Gene Expression Regulation , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/physiology
15.
Nat Immunol ; 13(10): 1010-9, 2012 Oct.
Article En | MEDLINE | ID: mdl-22922362

The transcription factor Foxp3 is indispensible for the differentiation and function of regulatory T cells (T(reg) cells). To gain insights into the molecular mechanisms of Foxp3-mediated gene expression, we purified Foxp3 complexes and explored their composition. Biochemical and mass-spectrometric analyses revealed that Foxp3 forms multiprotein complexes of 400-800 kDa or larger and identified 361 associated proteins, ∼30% of which were transcription related. Foxp3 directly regulated expression of a large proportion of the genes encoding its cofactors. Some transcription factor partners of Foxp3 facilitated its expression. Functional analysis of the cooperation of Foxp3 with one such partner, GATA-3, provided additional evidence for a network of transcriptional regulation afforded by Foxp3 and its associates to control distinct aspects of T(reg) cell biology.


Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Gene Regulatory Networks , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation , Forkhead Transcription Factors/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Humans , Mice , Mice, Transgenic , Protein Structure, Tertiary , Proteomics
16.
Immunity ; 34(4): 554-65, 2011 Apr 22.
Article En | MEDLINE | ID: mdl-21511184

T helper 17 (Th17) cells are important for host defense against extracellular microorganisms. However, they are also implicated in autoimmune and chronic inflammatory diseases, and as such need to be tightly regulated. The mechanisms that directly control committed pathogenic Th17 cells in vivo remain unclear. We showed here that IL-17A-producing CD4+ T cells expressed interleukin-10 receptor α (IL-10Rα) in vivo. Importantly, T cell-specific blockade of IL-10 signaling led to a selective increase of IL-17A+IFN-γ⁻ (Th17) and IL-17A+IFN-γ+ (Th17+Th1) CD4+ T cells during intestinal inflammation in the small intestine. CD4+Foxp3⁻ IL-10-producing (Tr1) cells and CD4+Foxp3+ regulatory (Treg) cells were able to control Th17 and Th17+Th1 cells in an IL-10-dependent manner in vivo. Lastly, IL-10 treatment of mice with established colitis decreased Th17 and Th17+Th1 cell frequencies via direct signaling in T cells. Thus, IL-10 signaling directly suppresses Th17 and Th17+Th1 cells.


CD4-Positive T-Lymphocytes/immunology , Interleukin-10 Receptor alpha Subunit/immunology , Interleukin-10/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Colitis/immunology , Colitis/pathology , Disease Progression , Forkhead Transcription Factors/immunology , Interferon-gamma/immunology , Interleukin-10/metabolism , Interleukin-10 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology , Signal Transduction , Th17 Cells/cytology , Th17 Cells/metabolism
17.
Immunity ; 34(4): 566-78, 2011 Apr 22.
Article En | MEDLINE | ID: mdl-21511185

Effector CD4+ T cell subsets, whose differentiation is facilitated by distinct cytokine cues, amplify the corresponding type of inflammatory response. Regulatory T (Treg) cells integrate environmental cues to suppress particular types of inflammation. In this regard, STAT3, a transcription factor essential for T helper 17 (Th17) cell differentiation, is necessary for Treg cell-mediated control of Th17 cell responses. Here, we showed that anti-inflammatory interleukin-10 (IL-10), and not proinflammatory IL-6 and IL-23 cytokine signaling, endowed Treg cells with the ability to suppress pathogenic Th17 cell responses. Ablation of the IL-10 receptor in Treg cells resulted in selective dysregulation of Th17 cell responses and colitis similar to that observed in mice harboring STAT3-deficient Treg cells. Thus, Treg cells limit Th17 cell inflammation by serving as principal amplifiers of negative regulatory circuits operating in immune effector cells.


Colitis/immunology , Interleukin-10/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Lineage , Colitis/pathology , Interleukin-10/metabolism , Mice , Mice, Knockout , Phosphorylation , Receptors, Interleukin-10/deficiency , Receptors, Interleukin-10/immunology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/cytology
18.
Cell ; 142(6): 914-29, 2010 Sep 17.
Article En | MEDLINE | ID: mdl-20850013

Foxp3(+) regulatory T (Treg) cells maintain immune homeostasis by limiting different types of inflammatory responses. Here, we report that miR-146a, one of the miRNAs prevalently expressed in Treg cells, is critical for their suppressor function. The deficiency of miR-146a in Treg cells resulted in a breakdown of immunological tolerance manifested in fatal IFNγ-dependent immune-mediated lesions in a variety of organs. This was likely due to augmented expression and activation of signal transducer and activator transcription 1 (Stat1), a direct target of miR-146a. Likewise, heightened Stat1 activation in Treg cells subjected to a selective ablation of SOCS1, a key negative regulator of Stat1 phosphorylation downstream of the IFNγ receptor, was associated with analogous Th1-mediated pathology. Our results suggest that specific aspects of Treg suppressor function are controlled by a single miRNA and that an optimal range of Stat1 activation is important for Treg-mediated control of Th1 responses and associated autoimmunity.


MicroRNAs/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Interferon-gamma/immunology , Mice , Mice, Knockout , MicroRNAs/genetics , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
19.
Nature ; 463(7282): 808-12, 2010 Feb 11.
Article En | MEDLINE | ID: mdl-20072126

Immune homeostasis is dependent on tight control over the size of a population of regulatory T (T(reg)) cells capable of suppressing over-exuberant immune responses. The T(reg) cell subset is comprised of cells that commit to the T(reg) lineage by upregulating the transcription factor Foxp3 either in the thymus (tT(reg)) or in the periphery (iT(reg)). Considering a central role for Foxp3 in T(reg) cell differentiation and function, we proposed that conserved non-coding DNA sequence (CNS) elements at the Foxp3 locus encode information defining the size, composition and stability of the T(reg) cell population. Here we describe the function of three Foxp3 CNS elements (CNS1-3) in T(reg) cell fate determination in mice. The pioneer element CNS3, which acts to potently increase the frequency of T(reg) cells generated in the thymus and the periphery, binds c-Rel in in vitro assays. In contrast, CNS1, which contains a TGF-beta-NFAT response element, is superfluous for tT(reg) cell differentiation, but has a prominent role in iT(reg) cell generation in gut-associated lymphoid tissues. CNS2, although dispensable for Foxp3 induction, is required for Foxp3 expression in the progeny of dividing T(reg) cells. Foxp3 binds to CNS2 in a Cbf-beta-Runx1 and CpG DNA demethylation-dependent manner, suggesting that Foxp3 recruitment to this 'cellular memory module' facilitates the heritable maintenance of the active state of the Foxp3 locus and, therefore, T(reg) lineage stability. Together, our studies demonstrate that the composition, size and maintenance of the T(reg) cell population are controlled by Foxp3 CNS elements engaged in response to distinct cell-extrinsic or -intrinsic cues.


Cell Lineage/genetics , Conserved Sequence/genetics , Forkhead Transcription Factors/genetics , Regulatory Sequences, Nucleic Acid/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation , Chromatin Assembly and Disassembly , CpG Islands/genetics , DNA Methylation , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-rel/metabolism , Response Elements/genetics , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
20.
J Immunol ; 183(12): 7631-4, 2009 Dec 15.
Article En | MEDLINE | ID: mdl-19923467

We have recently described two independent mouse models in which the administration of diphtheria toxin (DT) leads to specific depletion of regulatory T cells (Tregs) due to expression of DT receptor-enhanced GFP under the control of the Foxp3 promoter. Both mouse models develop severe autoimmune disorders when Foxp3(+) Tregs are depleted. Those findings were challenged in a recent study published in this journal suggesting the expression of Foxp3 in epithelial cells as the cause for disease development. By using genetic, cellular, and immunohistochemical approaches, we do not find evidence for Foxp3-expression in nonhematopoietic cells. DT injection does not lead to a loss of epithelial integrity in our Foxp3-DTR models. Instead, Foxp3 expression is Treg-specific and ablation of Foxp3(+) Tregs leads to the induction of fatal autoimmune disorders. Autoimmunity can be reversed by the adoptive transfer of Tregs into depleted hosts, and the transfer of Foxp3-deficient bone marrow into T cell-deficient irradiated recipients leads to full-blown disease development.


Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Targeting , Lymphocyte Depletion , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/pathology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Forkhead Transcription Factors/biosynthesis , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prostate/immunology , Prostate/metabolism , Prostate/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology
...