Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Immunol ; 205(10): 2667-2678, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33055282

ABSTRACT

IL-2 is a critical regulator of immune homeostasis through its impact on both regulatory T (Treg) and effector T cells. However, the precise role of IL-2 in the maintenance and function of Treg cells in the adult peripheral immune system remains unclear. In this study, we report that neutralization of IL-2 in mice abrogated all IL-2R signaling in Treg cells, but was well tolerated and only gradually impacted Treg cell function and immune homeostasis. By contrast, despite substantially reduced IL-2 sensitivity, Treg cells maintained selective IL-2 signaling and prevented immune dysregulation following treatment with the inhibitory anti-CD25 Ab PC61. Reduction of Treg cells with a depleting version of the same CD25 Ab permitted CD8+ effector T cell proliferation before progressing to more widespread immune dysregulation. Thus, despite severely curtailed CD25 expression and function, Treg cells retain selective access to IL-2 that supports their anti-inflammatory functions in vivo. Ab-mediated targeting of CD25 is being actively pursued for treatment of autoimmune disease and prevention of allograft rejection, and our findings help inform therapeutic manipulation and design for optimal patient outcomes.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Proliferation , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immune Tolerance/drug effects , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Models, Animal , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
2.
J Immunol ; 194(8): 3567-82, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25786692

ABSTRACT

The TNF family cytokine TL1A (Tnfsf15) costimulates T cells and type 2 innate lymphocytes (ILC2) through its receptor DR3 (Tnfrsf25). DR3-deficient mice have reduced T cell accumulation at the site of inflammation and reduced ILC2-dependent immune responses in a number of models of autoimmune and allergic diseases. In allergic lung disease models, immunopathology and local Th2 and ILC2 accumulation is reduced in DR3-deficient mice despite normal systemic priming of Th2 responses and generation of T cells secreting IL-13 and IL-4, prompting the question of whether TL1A promotes the development of other T cell subsets that secrete cytokines to drive allergic disease. In this study, we find that TL1A potently promotes generation of murine T cells producing IL-9 (Th9) by signaling through DR3 in a cell-intrinsic manner. TL1A enhances Th9 differentiation through an IL-2 and STAT5-dependent mechanism, unlike the TNF-family member OX40, which promotes Th9 through IL-4 and STAT6. Th9 differentiated in the presence of TL1A are more pathogenic, and endogenous TL1A signaling through DR3 on T cells is required for maximal pathology and IL-9 production in allergic lung inflammation. Taken together, these data identify TL1A-DR3 interactions as a novel pathway that promotes Th9 differentiation and pathogenicity. TL1A may be a potential therapeutic target in diseases dependent on IL-9.


Subject(s)
Asthma/immunology , Cell Differentiation/immunology , Interleukin-9/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Asthma/genetics , Asthma/pathology , Cell Differentiation/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-9/genetics , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
4.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33881452

ABSTRACT

A unique population of Foxp3+ regulatory T cells (TRs) resides in visceral adipose tissue (VAT) that regulates adipose inflammation and helps preserve insulin sensitivity. Inducible T cell co-stimulator (ICOS) is highly expressed on effector (e)TRs that migrate to nonlymphoid tissues, and contributes to their maintenance and function in models of autoimmunity. In this study, we report an unexpected cell-intrinsic role for ICOS expression and downstream phosphoinositide 3-kinase (PI3K) signaling in limiting the abundance, VAT-associated phenotype, and function of TRs specifically in VAT. Icos-/- mice and mice expressing a knock-in form of ICOS that cannot activate PI3K had increased VAT-TR abundance and elevated expression of canonical VAT-TR markers. Loss of ICOS signaling facilitated enhanced accumulation of TRs to VAT associated with elevated CCR3 expression, and resulted in reduced adipose inflammation and heightened insulin sensitivity in the context of a high-fat diet. Thus, we have uncovered a new and surprising molecular pathway that regulates VAT-TR accumulation and function.


Subject(s)
Adipose Tissue/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Diet, High-Fat/methods , Female , Forkhead Transcription Factors/immunology , Inflammation/immunology , Insulin/immunology , Insulin Resistance/immunology , Intra-Abdominal Fat/immunology , Male , Mice , Obesity/immunology , Phosphatidylinositol 3-Kinases/immunology
5.
Nat Commun ; 7: 10857, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26936133

ABSTRACT

Th9 cells produce interleukin (IL)-9, a cytokine implicated in allergic asthma and autoimmunity. Here we show that Itk, a mediator of T cell receptor signalling required for Th2 immune responses and the development of asthma, is a positive regulator of Th9 differentiation. In a model of allergic lung disease, Itk-deficient mice show reduced pulmonary inflammation and IL-9 production by T cells and innate lymphoid type 2 cells (ILC2), despite normal early induction of ILC2s. In vitro, Itk(-/-) CD4(+) T cells do not produce IL-9 and have reduced levels of IRF4 (Interferon Regulator Factor 4), a critical transcription factor for effector T cell function. Both IL-9 and IRF4 expression are rescued by either IL-2 or constitutively active STAT5, but not NFATc1. STAT5 binds the Irf4 promoter, demonstrating one mechanism by which IL-2 rescues weakly activated T cells. Itk inhibition also reduces IL-9 expression by human T cells, implicating ITK as a key regulator of Th9 induction.


Subject(s)
Cell Differentiation/physiology , Interferon Regulatory Factors/metabolism , Interleukin-2/metabolism , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes , Female , Gene Expression Regulation/physiology , Interferon Regulatory Factors/genetics , Interleukin-2/genetics , Lung Diseases/chemically induced , Male , Mice , Mice, Knockout , Papain/toxicity , Protein Kinases/genetics , Protein-Tyrosine Kinases/genetics
6.
J Leukoc Biol ; 98(3): 333-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26188076

ABSTRACT

Originally described in 2002 as a T cell-costimulatory cytokine, the tumor necrosis factor family member TNF-like factor 1A (TL1A), encoded by the TNFSF15 gene, has since been found to affect multiple cell lineages through its receptor, death receptor 3 (DR3, encoded by TNFRSF25) with distinct cell-type effects. Genetic deficiency or blockade of TL1A-DR3 has defined a number of disease states that depend on this cytokine-receptor pair, whereas excess TL1A leads to allergic gastrointestinal inflammation through stimulation of group 2 innate lymphoid cells. Noncoding variants in the TL1A locus are associated with susceptibility to inflammatory bowel disease and leprosy, predicting that the level of TL1A expression may influence host defense and the development of autoimmune and inflammatory diseases.


Subject(s)
Autoimmune Diseases/immunology , Receptors, Death Domain/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Animals , Autoimmune Diseases/pathology , Disease Models, Animal , Humans , Lymphocyte Activation/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL