Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37440178

ABSTRACT

Early-life cues shape the immune system during adulthood. However, early-life signaling pathways and their temporal functions are not well understood. Herein, we demonstrate that the cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1/2), which are E3 ubiquitin ligases, sustain interleukin (IL)-17-producing γ δ T cells (γδT17) and group 3 innate lymphoid cells (ILC3) during late neonatal and prepubescent life. We show that cell-intrinsic deficiency of cIAP1/2 at 3-4 wk of life leads to downregulation of the transcription factors cMAF and RORγt and failure to enter the cell cycle, followed by progressive loss of γδT17 cells and ILC3 during aging. Mice deficient in cIAP1/2 have severely reduced γδT17 cells and ILC3, present with suboptimal γδT17 responses in the skin, lack intestinal isolated lymphoid follicles, and cannot control intestinal bacterial infection. Mechanistically, these effects appear to be dependent on overt activation of the non-canonical NF-κB pathway. Our data identify cIAP1/2 as early-life molecular switches that establish effective type 3 immunity during aging.


Subject(s)
Immunity, Innate , Ubiquitin , Mice , Animals , Lymphocytes , Interleukins/metabolism , Aging
2.
Eur J Immunol ; 51(12): 3186-3193, 2021 12.
Article in English | MEDLINE | ID: mdl-34624928

ABSTRACT

Interleukin (IL)-17-producing gamma delta (γδ) T (γδT17) cells are an essential part of innate type 3 immunity against numerous pathogens. At the same time, a large body of evidence from mouse models and human clinical studies suggests that γδT17 cells contribute to the pathogenesis of many inflammatory diseases as well as cancer. It is therefore relevant to elucidate their immunobiology in detail and identify molecules and pathways that can regulate their function. Herein, we investigated the importance of the type I interferon (IFN) signaling system in γδT17 homeostasis and activation. We found that the IFN alpha receptor 1 (IFNAR1) was critical to maintain their normal homeostasis and to promote their activation during cutaneous inflammation. However, this did not require γδT17-intrinsic expression of IFNAR1. In contrast, expression of IFNAR1 by γδT17 cells was required in order to suppress IL-17 production during viral infection. Our data delineate direct from indirect IFNAR1 signaling and reveal an important immunoregulatory role for both tonic and inducible type I IFN in γδT17 cells.


Subject(s)
Interferon Type I/immunology , Lymphocyte Activation , Receptor, Interferon alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Interferon Type I/genetics , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction/genetics
3.
Eur J Immunol ; 51(9): 2164-2177, 2021 09.
Article in English | MEDLINE | ID: mdl-34224140

ABSTRACT

T cells of the gamma delta (γδ) lineage are evolutionary conserved from jawless to cartilaginous and bony fish to mammals and represent the "swiss army knife" of the immune system capable of antigen-dependent or independent responses, memory, antigen presentation, regulation of other lymphocytes, tissue homeostasis, and mucosal barrier maintenance, to list a few. Over the last 10 years, γδ T cells that produce the cytokine IL-17 (γδT17) have taken a leading position in our understanding of how our immune system battles infection, inflicts tissue damage during inflammation, and gets rewired by the tumor microenvironment. A lot of what we know about γδT17 cells stems from mouse models, however, increasing evidence implicates these cells in numerous human diseases. Herein, we aim to give an overview of the most common mouse models that have been used to study the role of γδT17 cells in infection, inflammation, and cancer, while at the same time we will evaluate evidence for their importance in humans. We hope and believe that in the next 10 years, means to take advantage of the protective and destructive properties of γδ T and in particular γδT17 cells will be part of our standard immunotherapy toolkit.


Subject(s)
Interleukin-17/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Th17 Cells/immunology , Animals , Autoimmune Diseases/immunology , Bacterial Infections/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Mice , Mycoses/immunology , Tumor Microenvironment/immunology , Virus Diseases/immunology
4.
Eur J Immunol ; 51(8): 2097-2099, 2021 08.
Article in English | MEDLINE | ID: mdl-33960415

ABSTRACT

SMAC antagonization of cIAP1/2 in TH 17 cells upregulates cell adhesion and cytoskeleton genes through the NIK-RelB and p52 axis. SMAC also increases the homotypic interactions of TH 17 cells through a non-canonical NF-κB- and integrin-mediated mechanism resulting in increased ability of TH 17 cells to withstand shear stress.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , Th17 Cells/metabolism , Baculoviral IAP Repeat-Containing 3 Protein/antagonists & inhibitors , Cell Adhesion/physiology , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lymphocyte Activation/physiology
5.
J Clin Invest ; 130(5): 2496-2508, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32281944

ABSTRACT

IL-17-producing RORγt+ γδ T cells (γδT17 cells) are innate lymphocytes that participate in type 3 immune responses during infection and inflammation. Herein, we show that γδT17 cells rapidly proliferate within neonatal lymph nodes and gut, where, upon entry, they upregulate T-bet and coexpress IL-17, IL-22, and IFN-γ in a STAT3- and retinoic acid-dependent manner. Neonatal expansion was halted in mice conditionally deficient in STAT5, and its loss resulted in γδT17 cell depletion from all adult organs. Hyperactive STAT5 mutant mice showed that the STAT5A homolog had a dominant role over STAT5B in promoting γδT17 cell expansion and downregulating gut-associated T-bet. In contrast, STAT5B preferentially expanded IFN-γ-producing γδ populations, implying a previously unknown differential role of STAT5 gene products in lymphocyte lineage regulation. Importantly, mice lacking γδT17 cells as a result of STAT5 deficiency displayed a profound resistance to experimental autoimmune encephalomyelitis. Our data identify that the neonatal microenvironment in combination with STAT5 is critical for post-thymic γδT17 development and tissue-specific imprinting, which is essential for infection and autoimmunity.


Subject(s)
Cellular Microenvironment/immunology , Immunity, Innate , Intestines/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , STAT5 Transcription Factor/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn , Cellular Microenvironment/genetics , Cytokines/genetics , Cytokines/immunology , Intestines/cytology , Mice , Mice, Transgenic , Organ Specificity/genetics , Organ Specificity/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , T-Lymphocytes/cytology
6.
Eur J Immunol ; 50(6): 873-879, 2020 06.
Article in English | MEDLINE | ID: mdl-32092146

ABSTRACT

IL-17-producing gamma delta (γδT17) cells are innate lymphocytes critical for antibacterial protection at barrier surfaces such as the skin but also highly pathogenic during inflammation. It is therefore important to understand the cellular and molecular mechanisms that could counter-balance overt γδT17 cell activation. Immune checkpoint receptors (ICRs) deliver inhibitory signals to activated lymphocytes and have been implicated as negative regulators of mouse γδT17 cells. In this report, we investigated the cytokine signals that induce ICR expression on γδT17 cells and studied the in vivo role of the Src-homology-2 phosphatases 1 and 2 (SHP-1 and SHP-2) in the context of γδT17-induced psoriasis. We found that surface expression of ICRs can be induced by cytokines; however, SHP-1 or SHP-2 could not inhibit γδT17 responses. In this regard, conditional deletion of SHP-1, SHP-2, or both did no impact γδT17 cell development, expansion, cytokine production, or skin pathology.


Subject(s)
Lymphocyte Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Skin/immunology , Th17 Cells/immunology , Animals , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Skin/pathology , Th17 Cells/pathology
7.
EMBO Rep ; 20(11): e48647, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31549795

ABSTRACT

The transcription factors STAT3 and STAT4 are essential for lymphocyte differentiation and function. Interleukin (IL)-17 producing γδ T (γδT17) cells are innate lymphocytes important for anti-bacterial and inflammatory responses at barrier surfaces. Herein, we examine the role of STAT3 and STAT4 in regulating the homeostasis, activation, and pathogenicity of γδT17 cells. We show that STAT3 sustains γδT17 numbers in the skin but not in the lymph nodes, while STAT4 deficiency does not affect their homeostasis. Similarly, STAT3 but not STAT4 is essential for IL-23-induced IL-22 production by γδT17 cells. Concomitantly, mice lacking STAT3 expression in γδT17 cells develop significantly reduced psoriasis-like inflammation. STAT3-deficient γδT17 cells fail to expand and to upregulate IL-17A, IL-17F, and IL-22 in response to psoriatic stimuli. Although STAT4-deficient animals develop psoriasis-like disease, γδT17 cells in these mice are defective in IL-17F production. Collectively, our data demonstrate for the first time a critical role for STAT3 in orchestrating the homeostasis and pathogenicity of γδT17 cells and provide evidence for the requirement of STAT4 for optimal cytokine responses during inflammation.


Subject(s)
Dermatitis/etiology , Dermatitis/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , STAT3 Transcription Factor/metabolism , STAT4 Transcription Factor/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Biopsy , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Immunomodulation , Immunophenotyping , Inflammation Mediators/metabolism , Lymphocyte Count , Mice , Psoriasis/etiology , Psoriasis/metabolism
8.
Sci Signal ; 12(596)2019 08 27.
Article in English | MEDLINE | ID: mdl-31455723

ABSTRACT

Second mitochondria-derived activator of caspase (SMAC) mimetics (SMs) are selective antagonists of the inhibitor of apoptosis proteins (IAPs), which activate noncanonical NF-κB signaling and promote tumor cell death. Through gene expression analysis, we found that treatment of CD4+ T cells with SMs during T helper 17 (TH17) cell differentiation disrupted the balance between two antagonistic transcription factor modules. Moreover, proteomics analysis revealed that SMs altered the abundance of proteins associated with cell cycle, mitochondrial activity, and the balance between canonical and noncanonical NF-κB signaling. Whereas SMs inhibited interleukin-17 (IL-17) production and ameliorated TH17 cell-driven inflammation, they stimulated IL-22 secretion. Mechanistically, SM-mediated activation of NF-κB-inducing kinase (NIK) and the transcription factors RelB and p52 directly suppressed Il17a expression and IL-17A protein production, as well as the expression of a number of other immune genes. Induction of IL-22 production correlated with the NIK-dependent reduction in cMAF protein abundance and the enhanced activity of the aryl hydrocarbon receptor. Last, SMs also increased IL-9 and IL-13 production and, under competing conditions, favored the differentiation of naïve CD4+ T cells into TH2 cells rather than TH17 cells. These results demonstrate that SMs shape the gene expression and protein profiles of TH17 cells and inhibit TH17 cell-driven autoimmunity.


Subject(s)
Apoptosis Regulatory Proteins , Biomimetic Materials/pharmacology , Cell Differentiation/immunology , Gene Expression Regulation/drug effects , Mitochondrial Proteins , Protein Serine-Threonine Kinases/immunology , Th17 Cells/immunology , Animals , Gene Expression Regulation/immunology , Mice , Mice, Transgenic , Th17 Cells/cytology , Th2 Cells/cytology , Th2 Cells/immunology , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...