Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nat Immunol ; 11(11): 1057-62, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20935646

ABSTRACT

After being activated by antigen, helper T lymphocytes switch from a resting state to clonal expansion. This switch requires inactivation of the transcription factor Foxo1, a suppressor of proliferation expressed in resting helper T lymphocytes. In the early antigen-dependent phase of expansion, Foxo1 is inactivated by antigen receptor-mediated post-translational modifications. Here we show that in the late phase of expansion, Foxo1 was no longer post-translationally regulated but was inhibited post-transcriptionally by the interleukin 2 (IL-2)-induced microRNA miR-182. Specific inhibition of miR-182 in helper T lymphocytes limited their population expansion in vitro and in vivo. Our results demonstrate a central role for miR-182 in the physiological regulation of IL-2-driven helper T cell-mediated immune responses and open new therapeutic possibilities.


Subject(s)
Interleukin-2/immunology , MicroRNAs/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Arthritis/immunology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
J Immunol ; 202(8): 2229-2239, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30833348

ABSTRACT

T follicular helper (Tfh) cells are a specialized T cell subset that regulates the long-lived production of highly specific Abs by B cells during the germinal center (GC) reaction. However, the transcriptional network sustaining the Tfh cell phenotype and function is still incompletely understood. In this study, we identify the transcription factor Bach2 as a central negative regulator of Tfh cells. Ectopic overexpression of Bach2 in murine Tfh cells resulted in a rapid loss of their phenotype and subsequent breakdown of the GC response. Low Bach2 expression levels are required to maintain high expression of the signature cytokine IL-21, the coinhibitory receptor TIGIT and the transcriptional repressor Bcl-6. In stark contrast to the regulatory network in GC B cells, Bach2 in Tfh cells is not coexpressed with Bcl-6 at high levels to inhibit the antagonizing factor Blimp-1, but suppresses Bcl-6 by direct binding to the promoter. These data reveal that by replacing an activating complex of Batf and Irf-4 at the Bcl-6 promoter, Bach2 regulates the transcriptional network of Tfh cells in a different way, as in GC B cells.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Gene Expression Regulation/immunology , Germinal Center/immunology , Proto-Oncogene Proteins c-bcl-6 , T-Lymphocytes, Regulatory/immunology , Transcription, Genetic/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Germinal Center/cytology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukins/genetics , Interleukins/immunology , Mice , Mice, Knockout , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , T-Lymphocytes, Regulatory/cytology
3.
Eur J Immunol ; 42(8): 1981-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22730020

ABSTRACT

T-follicular helper (TFH) cells represent the subpopulation of CD4(+) T cells that provides help for antigen-specific B cells in the GC response. They are generated from naïve T cells during an immune response and are imprinted by their master transcription factor Bcl-6. It has been a long-standing question if TFH cells contribute to the CD4(+) memory pool after the GC response has been terminated. To answer this question, we sorted antigen-specific TFH and non-TFH effector cells from an ongoing GC response and transferred them into naïve mice. Without further signals via the TCR, transferred cells rapidly contracted with a small population of both TFH and non-TFH cells surviving as memory cells in peripheral lymphoid organs for at least 4 weeks in the absence of antigen. TFH cells strongly downregulated their signature genes Bcl-6, CXCR5, and PD-1 in the memory phase. Upon rechallenge with antigen they rapidly upregulated these markers again. An enhanced potential to produce IL-21, paired with higher expression of CXCR5 and lower expression of CCR7, should enable TFH memory cells to provide more efficient help for antigen-specific B cells than their non-TFH counterparts.


Subject(s)
Antigens, Differentiation/biosynthesis , Immunologic Memory , Proto-Oncogene Proteins c-bcl-6/biosynthesis , Receptors, CXCR5/biosynthesis , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Antigens, Differentiation/immunology , Cell Differentiation , Down-Regulation , Germinal Center/immunology , Interleukins/biosynthesis , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-bcl-6/immunology , Receptors, CCR7/biosynthesis , Receptors, CXCR5/immunology , Spleen/immunology , Up-Regulation
4.
Elife ; 52016 12 30.
Article in English | MEDLINE | ID: mdl-28035902

ABSTRACT

The IL-2/IL-2Ralpha (CD25) axis is of central importance for the interplay of effector and regulatory T cells. Nevertheless, the question how different antigen loads are translated into appropriate IL-2 production to ensure adequate responses against pathogens remains largely unexplored. Here we find that at single cell level, IL-2 is binary (digital) and CD25 is graded expressed whereas at population level both parameters show graded expression correlating with the antigen amount. Combining in vivo data with a mathematical model we demonstrate that only this binary IL-2 expression ensures a wide linear antigen response range for Teff and Treg cells under real spatiotemporal conditions. Furthermore, at low antigen concentrations binary IL-2 expression safeguards by its spatial distribution selective STAT5 activation only of closely adjacent Treg cells regardless of their antigen specificity. These data show that the mode of IL-2 secretion is critical to tailor the adaptive immune response to the antigen amount.


Subject(s)
Adoptive Transfer , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Gene Expression Regulation , Immunization , Immunophenotyping , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Ovalbumin/administration & dosage , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , Single-Cell Analysis , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/transplantation , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/transplantation , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
5.
J Exp Med ; 212(2): 217-33, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25646266

ABSTRACT

The co-stimulators ICOS (inducible T cell co-stimulator) and CD28 are both important for T follicular helper (TFH) cells, yet their individual contributions are unclear. Here, we show that each molecule plays an exclusive role at different stages of TFH cell development. While CD28 regulated early expression of the master transcription factor Bcl-6, ICOS co-stimulation was essential to maintain the phenotype by regulating the novel TFH transcription factor Klf2 via Foxo1. Klf2 directly binds to Cxcr5, Ccr7, Psgl-1, and S1pr1, and low levels of Klf2 were essential to maintain this typical TFH homing receptor pattern. Blocking ICOS resulted in relocation of fully developed TFH cells back to the T cell zone and reversion of their phenotype to non-TFH effector cells, which ultimately resulted in breakdown of the germinal center response. Our study describes for the first time the exclusive role of ICOS and its downstream signaling in the maintenance of TFH cells by controlling their anatomical localization in the B cell follicle.


Subject(s)
Gene Expression Regulation , Inducible T-Cell Co-Stimulator Protein/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Phenotype , T-Lymphocyte Subsets , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CD28 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Down-Regulation , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression , Germinal Center/immunology , Germinal Center/metabolism , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Protein Binding , Receptors, CCR7/metabolism , Receptors, CXCR5/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology
SELECTION OF CITATIONS
SEARCH DETAIL