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.
Cell Mol Immunol ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961265

ABSTRACT

Immune-mediated nephritis is a leading cause of acute kidney injury and chronic kidney disease. While the role of B cells and antibodies has been extensively investigated in the past, the advent of immune-checkpoint inhibitors has led to a reappraisal of the role of T cells in renal immunology. However, it remains elusive how T cells with specificity for renal autoantigens are activated and participate in immune-mediated nephritis. Here, we followed the fate and function of pathogen-activated autoreactive CD8 T cells that are specific for a renal autoantigen. We demonstrate that recently activated splenic CD8 T cells developed a hybrid phenotype in the context of renal autoantigen cross-presentation, combining hallmarks of activation and T cell dysfunction. While circulating memory T cells rapidly disappeared, tissue-resident memory T cells emerged and persisted within the kidney, orchestrating immune-mediated nephritis. Notably, T cells infiltrating kidneys of patients with interstitial nephritis also expressed key markers of tissue residency. This study unveils how a tissue-specific immune response can dissociate from its systemic counterpart driving a compartmentalized immune response in the kidneys of mice and man. Consequently, targeting tissue-resident memory T cells emerges as a promising strategy to control immune-mediated kidney disease.

2.
Cell Death Differ ; 31(7): 924-937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849575

ABSTRACT

Mitochondria react to infection with sub-lethal signals in the apoptosis pathway. Mitochondrial signals can be inflammatory but mechanisms are only partially understood. We show that activation of the caspase-activated DNase (CAD) mediates mitochondrial pro-inflammatory functions and substantially contributes to host defense against viral infection. In cells lacking CAD, the pro-inflammatory activity of sub-lethal signals was reduced. Experimental activation of CAD caused transient DNA-damage and a pronounced DNA damage response, involving major kinase signaling pathways, NF-κB and cGAS/STING, driving the production of interferon, cytokines/chemokines and attracting neutrophils. The transcriptional response to CAD-activation was reminiscent of the reaction to microbial infection. CAD-deficient cells had a diminished response to viral infection. Influenza virus infected CAD-deficient mice displayed reduced inflammation in lung tissue, higher viral titers and increased weight loss. Thus, CAD links the mitochondrial apoptosis system and cell death caspases to host defense. CAD-driven DNA damage is a physiological element of the inflammatory response to infection.


Subject(s)
DNA Damage , Inflammation , Mitochondria , Animals , Inflammation/pathology , Inflammation/metabolism , Mice , Mitochondria/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/metabolism , Mice, Inbred C57BL , Apoptosis , Humans , NF-kappa B/metabolism , Deoxyribonucleases/metabolism , Deoxyribonucleases/genetics , Mice, Knockout , Signal Transduction , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/deficiency , Nucleotidyltransferases
3.
Sci Adv ; 10(11): eadj2802, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489359

ABSTRACT

Development of T cells is controlled by the signal strength of the TCR. The scaffold protein kinase D-interacting substrate of 220 kilodalton (Kidins220) binds to the TCR; however, its role in T cell development was unknown. Here, we show that T cell-specific Kidins220 knockout (T-KO) mice have strongly reduced invariant natural killer T (iNKT) cell numbers and modest decreases in conventional T cells. Enhanced apoptosis due to increased TCR signaling in T-KO iNKT thymocytes of developmental stages 2 and 3 shows that Kidins220 down-regulates TCR signaling at these stages. scRNA-seq indicated that the transcription factor Aiolos is down-regulated in Kidins220-deficient iNKT cells. Analysis of an Aiolos KO demonstrated that Aiolos is a downstream effector of Kidins220 during iNKT cell development. In the periphery, T-KO iNKT cells show reduced TCR signaling upon stimulation with α-galactosylceramide, suggesting that Kidins220 promotes TCR signaling in peripheral iNKT cells. Thus, Kidins220 reduces or promotes signaling dependent on the iNKT cell developmental stage.


Subject(s)
Ikaros Transcription Factor , Membrane Proteins , Natural Killer T-Cells , Thymus Gland , Animals , Mice , Cell Differentiation , Gene Expression Regulation , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Membrane Proteins/metabolism , Ikaros Transcription Factor/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
4.
Front Immunol ; 14: 1058267, 2023.
Article in English | MEDLINE | ID: mdl-36756120

ABSTRACT

The T-box transcription factors T-bet and Eomesodermin regulate type 1 immune responses in innate and adaptive lymphocytes. T-bet is widely expressed in the immune system but was initially identified as the lineage-specifying transcription factor of Th1 CD4+ T cells, where it governs expression of the signature cytokine IFN- γ and represses alternative cell fates like Th2 and Th17. T-bet's paralog Eomes is less abundantly expressed and Eomes+ CD4+ T cells are mostly found in the context of persistent antigen exposure, like bone marrow transplantation, chronic infection or inflammation as well as malignant disorders. However, it has remained unresolved whether Eomes executes similar transcriptional activities as T-bet in CD4+ T cells. Here we use a novel genetic approach to show that Eomes expression in CD4+ T cells drives a distinct transcriptional program that shows only partial overlap with T-bet. We found that Eomes is sufficient to induce the expression of the immunoregulatory cytokine IL-10 and, together with T-bet, promotes a cytotoxic effector profile, including Prf1, Gzmb, Gzmk, Nkg7 and Ccl5, while repressing alternative cell fates. Our results demonstrate that Eomes+ CD4+ T cells, which are often found in the context of chronic antigen stimulation, are likely to be a unique CD4+ T cell subset that limits inflammation and immunopathology as well as eliminates antigen-presenting and malignant cells.


Subject(s)
Antineoplastic Agents , Interleukin-10 , Mice , Animals , Interleukin-10/genetics , Interferon-gamma/metabolism , T-Lymphocyte Subsets , Cytokines , Th17 Cells , Inflammation , T-Box Domain Proteins/genetics , Membrane Proteins
5.
PLoS Pathog ; 16(9): e1008870, 2020 09.
Article in English | MEDLINE | ID: mdl-32991634

ABSTRACT

The two T-box transcription factors T-bet and Eomesodermin (Eomes) are important regulators of cytotoxic lymphocytes (CTLs), such as activated CD8 T cells, which are essential in the fight against intracellular pathogens and tumors. Both transcription factors share a great degree of homology based on sequence analysis and as a result exert partial functional redundancy during viral infection. However, the actual degree of redundancy between T-bet and Eomes remains a matter of debate and is further confounded by their distinct spatiotemporal expression pattern in activated CD8 T cells. To directly investigate the functional overlap of these transcription factors, we generated a new mouse model in which Eomes expression is under the transcriptional control of the endogenous Tbx21 (encoding for T-bet) locus. Applying this model, we demonstrate that the induction of Eomes in lieu of T-bet cannot rescue T-bet deficiency in CD8 T cells during acute lymphocytic choriomeningitis virus (LCMV) infection. We found that the expression of Eomes instead of T-bet was not sufficient for early cell expansion or effector cell differentiation. Finally, we show that imposed expression of Eomes after acute viral infection promotes some features of exhaustion but must act in concert with other factors during chronic viral infection to establish all hallmarks of exhaustion. In summary, our results clearly underline the importance of T-bet in guiding canonical CTL development during acute viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , T-Box Domain Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Fetal Proteins/metabolism , Gene Expression Regulation/physiology , Interferon-gamma/metabolism , Mice, Transgenic
6.
Mucosal Immunol ; 13(2): 257-270, 2020 03.
Article in English | MEDLINE | ID: mdl-31712600

ABSTRACT

Natural intraepithelial lymphocytes (IELs) are thymus-derived adaptive immune cells, which are important contributors to intestinal immune homeostasis. Similar to other innate-like T cells, they are induced in the thymus through high-avidity interaction that would otherwise lead to clonal deletion in conventional CD4 and CD8 T cells. By applying single-cell RNA-sequencing (scRNA-seq) on a heterogeneous population of thymic CD4-CD8αß-TCRαß+NK1.1- IEL precursors (NK1.1- IELPs), we define a developmental trajectory that can be tracked based on the sequential expression of CD122 and T-bet. Moreover, we identify the Id proteins Id2 and Id3 as a novel regulator of IELP development and show that all NK1.1- IELPs progress through a PD-1 stage that precedes the induction of T-bet. The transition from PD-1 to T-bet is regulated by the transcription factor C-Myc, which has far reaching effects on cell cycle, energy metabolism, and the translational machinery during IELP development. In summary, our results provide a high-resolution molecular framework for thymic IEL development of NK1.1- IELPs and deepen our understanding of this still elusive cell type.


Subject(s)
Intraepithelial Lymphocytes/immunology , Precursor Cells, T-Lymphoid/immunology , Proto-Oncogene Proteins c-myc/metabolism , T-Box Domain Proteins/metabolism , Thymus Gland/immunology , Animals , Antigens, Ly/metabolism , Cells, Cultured , Gene Expression Regulation , Immunity, Innate , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , T-Box Domain Proteins/genetics
7.
Mucosal Immunol ; 11(2): 333-344, 2018 03.
Article in English | MEDLINE | ID: mdl-28745324

ABSTRACT

The intestine is a major immune organ with several specialized lymphoid structures and immune cells. Among these are thymus-derived natural intraepithelial lymphocytes (IELs) that lack expression of the classical co-receptors CD4 or CD8αß (double negative (DN)). Natural IELs are both αß+ and γδ+ T cells that play important roles in the maintenance of the epithelial barrier at steady state and during inflammation. The transcription factor T-bet is essential for the peripheral development of natural IELs, but its role during thymic development has remained less clear. Here we show that a T-bet gradient in DN TCRαß+NK1.1- thymocytes (IEL precursors (IELPs)) determines IEL fate in natural TCRαß+ IELs. Employing T-bet ZsGreen reporter mice in in vitro cultures and in vivo transfer experiments, we demonstrate that with increasing expression of T-bet, DN TCRαß+NK1.1- thymocytes are gradually restricted to a DN IEL fate. Furthermore, we show that the natural TCRαß+ IELs seed the intestine within the first month of life. This in turn is preceded by the appearance of T-bet- and T-bet+ IELPs that egress from the thymus in a sphingosine-1-phosphate (S1P)-dependent manner. In summary, the use of T-bet reporter mice has enabled us to identify and refine an immediate and clearly committed postselection precursor of natural TCRαß+ IELs.


Subject(s)
Intestines/immunology , Intraepithelial Lymphocytes/physiology , Precursor Cells, T-Lymphoid/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Animals , Cell Differentiation , Cells, Cultured , Clonal Selection, Antigen-Mediated , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
8.
Genesis ; 55(8)2017 08.
Article in English | MEDLINE | ID: mdl-28646547

ABSTRACT

The T-box transcription factor Eomes (also known as Tbr2) shows short-lived expression in various localized domains of the embryo, including epiblast cells during gastrulation and intermediate progenitor cells in the cerebral cortex. In these tissues Eomes fulfills crucial roles for lineage specification of progenitors. To directly observe Eomes-dependent cell lineages in the living embryo, we generated a novel dual-fluorescence reporter allele that expresses a membrane-bound tdTomato protein for investigation of cell morphology and a nuclear GFP for cell tracing. This allele recapitulates endogenous EOMES protein expression and is suitable for live imaging. We found that the allele can also be used as a short-to-medium-term lineage tracer, as GFP persists in cells longer than EOMES protein and marks Eomes-dependent lineages with a timeframe of days to weeks depending on the proliferation rate. In summary, we present a novel genetic tool for investigation of Eomes-dependent cell types by live imaging and lineage tracing.


Subject(s)
Cell Lineage , Cell Tracking/methods , Genes, Reporter , Genetic Engineering/methods , Optical Imaging/methods , T-Box Domain Proteins/genetics , Animals , Female , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , T-Box Domain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL