Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Immunity ; 56(7): 1631-1648.e10, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392737

ABSTRACT

CD137 (4-1BB)-activating receptor represents a promising cancer immunotherapeutic target. Yet, the cellular program driven by CD137 and its role in cancer immune surveillance remain unresolved. Using T cell-specific deletion and agonist antibodies, we found that CD137 modulates tumor infiltration of CD8+-exhausted T (Tex) cells expressing PD1, Lag-3, and Tim-3 inhibitory receptors. T cell-intrinsic, TCR-independent CD137 signaling stimulated the proliferation and the terminal differentiation of Tex precursor cells through a mechanism involving the RelA and cRel canonical NF-κB subunits and Tox-dependent chromatin remodeling. While Tex cell accumulation induced by prophylactic CD137 agonists favored tumor growth, anti-PD1 efficacy was improved with subsequent CD137 stimulation in pre-clinical mouse models. Better understanding of T cell exhaustion has crucial implications for the treatment of cancer and infectious diseases. Our results identify CD137 as a critical regulator of Tex cell expansion and differentiation that holds potential for broad therapeutic applications.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Cell Differentiation , Cell Proliferation , Receptors, Antigen, T-Cell
2.
Nat Immunol ; 17(2): 140-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26657003

ABSTRACT

Innate sensing of pathogens initiates inflammatory cytokine responses that need to be tightly controlled. We found here that after engagement of Toll-like receptors (TLRs) in myeloid cells, deficient sumoylation caused increased secretion of transcription factor NF-κB-dependent inflammatory cytokines and a massive type I interferon signature. In mice, diminished sumoylation conferred susceptibility to endotoxin shock and resistance to viral infection. Overproduction of several NF-κB-dependent inflammatory cytokines required expression of the type I interferon receptor, which identified type I interferon as a central sumoylation-controlled hub for inflammation. Mechanistically, the small ubiquitin-like modifier SUMO operated from a distal enhancer of the gene encoding interferon-ß (Ifnb1) to silence both basal and stimulus-induced activity of the Ifnb1 promoter. Therefore, sumoylation restrained inflammation by silencing Ifnb1 expression and by strictly suppressing an unanticipated priming by type I interferons of the TLR-induced production of inflammatory cytokines.


Subject(s)
Disease Resistance , Gene Expression Regulation , Immunity, Innate , Immunomodulation , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Sumoylation , Animals , Chromatin/genetics , Chromatin/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Disease Susceptibility , Enhancer Elements, Genetic , Gene Expression Profiling , Genetic Loci , Inflammation/virology , Inflammation Mediators/metabolism , Interferon-beta/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Protein Binding , Receptor, Interferon alpha-beta/metabolism , Regulatory Elements, Transcriptional , SUMO-1 Protein/metabolism , Shock, Septic/genetics , Shock, Septic/immunology , Shock, Septic/metabolism , Signal Transduction , Sumoylation/genetics , Sumoylation/immunology , Toll-Like Receptors/metabolism
3.
Immunity ; 50(3): 629-644.e8, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30737147

ABSTRACT

Upon activation, naive CD4+ T cells differentiate into distinct T cell subsets via processes reliant on epigenetically regulated, lineage-specific developmental programs. Here, we examined the function of the histone methyltransferase SETDB1 in T helper (Th) cell differentiation. Setdb1-/- naive CD4+ T cells exhibited exacerbated Th1 priming, and when exposed to a Th1-instructive signal, Setdb1-/- Th2 cells crossed lineage boundaries and acquired a Th1 phenotype. SETDB1 did not directly control Th1 gene promoter activity but relied instead on deposition of the repressive H3K9me3 mark at a restricted and cell-type-specific set of endogenous retroviruses (ERVs) located in the vicinity of genes involved in immune processes. Refined bioinformatic analyses suggest that these retrotransposons regulate Th1 gene cis-regulatory elements or act as Th1 gene enhancers. Thus, H3K9me3 deposition by SETDB1 ensures Th cell lineage integrity by repressing a repertoire of ERVs that have been exapted into cis-regulatory modules to shape and control the Th1 gene network.


Subject(s)
Cell Lineage/immunology , Endogenous Retroviruses/immunology , Histone Methyltransferases/immunology , Histone-Lysine N-Methyltransferase/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Female , Histones/immunology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/immunology , Th1 Cells/immunology , Th2 Cells/immunology
4.
Nat Immunol ; 16(6): 628-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939024

ABSTRACT

Most T lymphocytes, including regulatory T cells (Treg cells), differentiate in the thymus. The age-dependent involution of this organ leads to decreasing production of T cells. Here we found that the output of new Treg cells from the thymus decreased substantially more than that of conventional T cells. Peripheral mouse and human Treg cells recirculated back to the thymus, where they constituted a large proportion of the pool of Treg cells and displayed an activated and differentiated phenotype. In the thymus, the recirculating cells exerted their regulatory function by inhibiting interleukin 2 (IL-2)-dependent de novo differentiation of Treg cells. Thus, Treg cell development is controlled by a negative feedback loop in which mature progeny cells return to the thymus and restrain development of precursors of Treg cells.


Subject(s)
Precursor Cells, T-Lymphoid/physiology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/immunology , Aging/immunology , Animals , Blood Circulation , Cell Differentiation/genetics , Cells, Cultured , Child , Feedback, Physiological , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Nephrol Dial Transplant ; 39(3): 496-509, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-37697719

ABSTRACT

BACKGROUND: The role of macrophages in the development of rhabdomyolysis-induced acute kidney injury (RM-AKI) has been established, but an in-depth understanding of the changes in the immune landscape could help to improve targeted strategies. Whereas senescence is usually associated with chronic kidney processes, we also wished to explore whether senescence could also occur in AKI and whether senolytics could act on immune cells. METHODS: Single-cell RNA sequencing was used in the murine glycerol-induced RM-AKI model to dissect the transcriptomic characteristics of CD45+ live cells sorted from kidneys 2 days after injury. Public datasets from murine AKI models were reanalysed to explore cellular senescence signature in tubular epithelial cells (TECs). A combination of senolytics (dasatinib and quercetin, DQ) was administered to mice exposed or not to RM-AKI. RESULTS: Unsupervised clustering of nearly 17 000 single-cell transcriptomes identified seven known immune cell clusters. Sub-clustering of the mononuclear phagocyte cells revealed nine distinct cell sub-populations differently modified with RM. One macrophage cluster was particularly interesting since it behaved as a critical node in a trajectory connecting one major histocompatibility complex class IIhigh (MHCIIhigh) cluster only present in Control to two MHCIIlow clusters only present in RM-AKI. This critical cluster expressed a senescence gene signature, that was very different from that of the TECs. Senolytic DQ treatment blocked the switch from a F4/80highCD11blow to F4/80lowCD11bhigh phenotype, which correlated with prolonged nephroprotection in RM-AKI. CONCLUSIONS: Single-cell RNA sequencing unmasked novel transitional macrophage subpopulation associated with RM-AKI characterized by the activation of cellular senescence processes. This work provides a proof-of-concept that senolytics nephroprotective effects may rely, at least in part, on subtle immune modulation.


Subject(s)
Acute Kidney Injury , Rhabdomyolysis , Mice , Animals , Senotherapeutics , Acute Kidney Injury/etiology , Acute Kidney Injury/complications , Kidney , Rhabdomyolysis/complications , Rhabdomyolysis/drug therapy , Sequence Analysis, RNA
6.
Eur J Immunol ; 51(3): 580-593, 2021 03.
Article in English | MEDLINE | ID: mdl-32730634

ABSTRACT

Regulatory T lymphocytes (Treg) play a vital role in the protection of the organism against autoimmune pathology. It is therefore paradoxical that comparatively large numbers of Treg were found in the thymus of type I diabetes-prone NOD mice. The Treg population in the thymus is composed of newly developing cells and cells that had recirculated from the periphery back to the thymus. We here demonstrate that exceptionally large numbers of Treg develop in the thymus of young, but not adult, NOD mice. Once emigrated from the thymus, an unusually large proportion of these Treg is activated in the periphery, which causes a particularly abundant accumulation of recirculating Treg in the thymus. These cells then rapidly inhibit de novo development of Treg. The proportions of developing Treg thus reach levels similar to or lower than those found in most other, type 1 diabetes-resistant, inbred mouse strains. Thus, in adult NOD mice the particularly large Treg-niche is actually composed of mostly recirculating cells and only few newly developing Treg.


Subject(s)
T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/immunology , Male , Mice , Mice, Inbred NOD
7.
Proc Natl Acad Sci U S A ; 116(51): 25839-25849, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776254

ABSTRACT

Naive CD4+ T lymphocytes differentiate into different effector types, including helper and regulatory cells (Th and Treg, respectively). Heritable gene expression programs that define these effector types are established during differentiation, but little is known about the epigenetic mechanisms that install and maintain these programs. Here, we use mice defective for different components of heterochromatin-dependent gene silencing to investigate the epigenetic control of CD4+ T cell plasticity. We show that, upon T cell receptor (TCR) engagement, naive and regulatory T cells defective for TRIM28 (an epigenetic adaptor for histone binding modules) or for heterochromatin protein 1 ß and γ isoforms (HP1ß/γ, 2 histone-binding factors involved in gene silencing) fail to effectively signal through the PI3K-AKT-mTOR axis and switch to glycolysis. While differentiation of naive TRIM28-/- T cells into cytokine-producing effector T cells is impaired, resulting in reduced induction of autoimmune colitis, TRIM28-/- regulatory T cells also fail to expand in vivo and to suppress autoimmunity effectively. Using a combination of transcriptome and chromatin immunoprecipitation-sequencing (ChIP-seq) analyses for H3K9me3, H3K9Ac, and RNA polymerase II, we show that reduced effector differentiation correlates with impaired transcriptional silencing at distal regulatory regions of a defined set of Treg-associated genes, including, for example, NRP1 or Snai3. We conclude that TRIM28 and HP1ß/γ control metabolic reprograming through epigenetic silencing of a defined set of Treg-characteristic genes, thus allowing effective T cell expansion and differentiation into helper and regulatory phenotypes.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic/physiology , T-Lymphocytes/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Animals , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Plasticity/physiology , Cellular Reprogramming/genetics , Chromobox Protein Homolog 5 , Colon/pathology , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Silencing , Histones/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcriptome , Tripartite Motif-Containing Protein 28/genetics
8.
Immunology ; 148(2): 187-96, 2016 06.
Article in English | MEDLINE | ID: mdl-26924728

ABSTRACT

Regulatory T (Treg) lymphocytes play a central role in the control of immune responses and so maintain immune tolerance and homeostasis. In mice, expression of the CD8 co-receptor and low levels of the co-stimulatory molecule CD28 characterizes a Treg cell population that exerts potent suppressive function in vitro and efficiently controls experimental immunopathology in vivo. It has remained unclear if CD8(+) CD28(low) Treg cells develop in the thymus or represent a population of chronically activated conventional T cells differentiating into Treg cells in the periphery, as suggested by their CD28(low) phenotype. We demonstrate that functional CD8(+) CD28(low) Treg cells are present in the thymus and that these cells develop locally and are not recirculating from the periphery. Differentiation of CD8(+) CD28(low) Treg cells requires MHC class I expression on radioresistant but not on haematopoietic thymic stromal cells. In contrast to other Treg cells, CD8(+) CD28(low) Treg cells develop simultaneously with CD8(+) CD28(high) conventional T cells. We also identified a novel homologous naive CD8(+) CD28(low) T-cell population with immunosuppressive properties in human blood and thymus. Combined, our data demonstrate that CD8(+) CD28(low) cells can develop in the thymus of mice and suggest that the same is true in humans.


Subject(s)
T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/physiology , Animals , CD28 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Humans , Immune Tolerance , Immunosuppression Therapy , Mice , Mice, Inbred C57BL
9.
Nature ; 458(7240): 899-903, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19219027

ABSTRACT

Injury or impaired clearance of apoptotic cells leads to the pathological accumulation of necrotic corpses, which induce an inflammatory response that initiates tissue repair. In addition, antigens present in necrotic cells can sometimes provoke a specific immune response and it has been argued that necrosis could explain adaptive immunity in seemingly infection-free situations, such as after allograft transplantation or in spontaneous and therapy-induced tumour rejection. In the mouse, the CD8alpha+ subset of dendritic cells phagocytoses dead cell remnants and cross-primes CD8+ T cells against cell-associated antigens. Here we show that CD8alpha+ dendritic cells use CLEC9A (also known as DNGR-1), a recently-characterized C-type lectin, to recognize a preformed signal that is exposed on necrotic cells. Loss or blockade of CLEC9A does not impair the uptake of necrotic cell material by CD8+ dendritic cells, but specifically reduces cross-presentation of dead-cell-associated antigens in vitro and decreases the immunogenicity of necrotic cells in vivo. The function of CLEC9A requires a key tyrosine residue in its intracellular tail that allows the recruitment and activation of the tyrosine kinase SYK, which is also essential for cross-presentation of dead-cell-associated antigens. Thus, CLEC9A functions as a SYK-coupled C-type lectin receptor to mediate sensing of necrosis by the principal dendritic-cell subset involved in regulating cross-priming to cell-associated antigens.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Necrosis/immunology , Necrosis/metabolism , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , Animals , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Cross-Priming/immunology , Humans , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Ligands , Mice , Phagocytosis , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Mitogen/genetics , Signal Transduction
10.
Front Immunol ; 15: 1358459, 2024.
Article in English | MEDLINE | ID: mdl-38404576

ABSTRACT

Regulatory T lymphocytes expressing the transcription factor Foxp3 (Tregs) play an important role in the prevention of autoimmune diseases and other immunopathologies. Aberrations in Treg-mediated immunosuppression are therefore thought to be involved in the development of autoimmune pathologies, but few have been documented. Recent reports indicated a central role for Tregs developing during the neonatal period in the prevention of autoimmune pathology. We therefore investigated the development of Tregs in neonatal NOD mice, an important animal model for autoimmune type 1 diabetes. Surprisingly, we found that, as compared with seven other commonly studied inbred mouse strains, in neonatal NOD mice, exceptionally large proportions of developing Tregs express high levels of GITR and PD-1. The latter phenotype was previously associated with high Treg autoreactivity in C57BL/6 mice, which we here confirm for NOD animals. The proportions of newly developing GITRhighPD-1+ Tregs rapidly drop during the first week of age. A genome-wide genetic screen indicated the involvement of several diabetes susceptibility loci in this trait. Analysis of a congenic mouse strain confirmed that Idd5 contributes to the genetic control of GITRhighPD-1+ Treg development in neonates. Our data thus demonstrate an intriguing and paradoxical correlation between an idiosyncrasy in Treg development in NOD mice and their susceptibility to type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Animals , Diabetes Mellitus, Type 1/genetics , Mice, Inbred NOD , T-Lymphocytes, Regulatory , Programmed Cell Death 1 Receptor/genetics , Mice, Inbred C57BL , Transcription Factors
11.
J Exp Med ; 204(6): 1487-501, 2007 Jun 11.
Article in English | MEDLINE | ID: mdl-17548522

ABSTRACT

Dendritic cell (DC) activation is a prerequisite for T cell priming. During infection, activation can ensue from signaling via pattern-recognition receptors after contact with pathogens or infected cells. Alternatively, it has been proposed that DCs can be activated indirectly by signals produced by infected tissues. To address the contribution of tissue-derived signals, we measured DC activation in a model in which radioresistant cells can or cannot respond to lipopolysaccharide (LPS). We report that recognition of LPS by the radioresistant compartment is sufficient to induce local and systemic inflammation characterized by high circulating levels of tumor necrosis factor (TNF) alpha, interleukin (IL) 1beta, IL-6, and CC chemokine ligand 2. However, this is not sufficient to activate DCs, whether measured by migration, gene expression, phenotypic, or functional criteria, or to render DC refractory to subsequent stimulation with CpG-containing DNA. Similarly, acute or chronic exposure to proinflammatory cytokines such as TNF-alpha +/- interferon alpha/beta has marginal effects on DC phenotype in vivo when compared with LPS. In addition, DC activation and migration induced by LPS is unimpaired when radioresistant cells cannot respond to the stimulus. Thus, inflammatory mediators originating from nonhematopoietic tissues and from radioresistant hematopoietic cells are neither sufficient nor required for DC activation in vivo.


Subject(s)
Dendritic Cells/immunology , Inflammation/immunology , Signal Transduction/immunology , Animals , Cell Movement/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Immunohistochemistry , Lipopolysaccharides , Mice , Mice, Mutant Strains , Models, Immunological , Receptors, Cytokine/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
12.
Immunol Rev ; 227(1): 234-47, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19120488

ABSTRACT

Pathogen invasion induces a rapid inflammatory response initiated through the recognition of pathogen-derived molecules by pattern recognition receptors (PRRs) expressed on both immune and non-immune cells. The initial wave of pro-inflammatory cytokines and chemokines limits pathogen spread and recruits and activates immune cells to eradicate the invaders. Dendritic cells (DCs) are responsible for initiating a subsequent phase of immunity, dominated by the action of pathogen-specific T and B cells. As for the early pro-inflammatory response, DC activation is triggered by PRR signals. These signals convert resting DCs into potent antigen-presenting cells capable of promoting the expansion and effector differentiation of naive pathogen-specific T cells. However, it has been argued that signals from PRRs are not a prerequisite for DC activation and that pro-inflammatory cytokines have the same effect. Although this may appear like an efficient way to expand the number of DCs that initiate adaptive immunity, evidence is accumulating that DCs activated indirectly by inflammatory cytokines are unable to induce functional T-cell responses. Here, we review the differences between PRR-triggered and cytokine-induced DC activation and speculate on a potential role for DCs activated by inflammatory signals in tolerance induction rather than immunity.


Subject(s)
Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Cellular , Receptors, Pattern Recognition/metabolism , Signal Transduction/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Animals , Cell Differentiation/immunology , Cross-Priming/immunology , Cytokines/genetics , Cytokines/immunology , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Dendritic Cells/pathology , Immune Tolerance/immunology , Immunity, Innate , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Mice , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
13.
Front Immunol ; 13: 965303, 2022.
Article in English | MEDLINE | ID: mdl-36159793

ABSTRACT

Development of Foxp3-expressing regulatory T-lymphocytes (Treg) in the thymus is controlled by signals delivered in T-cell precursors via the TCR, co-stimulatory receptors, and cytokine receptors. In absence of IL-2, IL-15 or their receptors, fewer Treg apparently develop in the thymus. However, it was recently shown that a substantial part of thymic Treg are cells that had recirculated from the periphery back to the thymus, troubling interpretation of these results. We therefore reassessed the involvement of IL-2 and IL-15 in the development of Treg, taking into account Treg-recirculation. At the age of three weeks, when in wt and IL-15-deficient (but not in IL-2-deficient) mice substantial amounts of recirculating Treg are present in the thymus, we found similarly reduced proportions of newly developed Treg in absence of IL-2 or IL-15, and in absence of both cytokines even less Treg developed. In neonates, when practically no recirculating Treg were found in the thymus, the absence of IL-2 led to substantially more reduced Treg-development than deficiency in IL-15. IL-2 but not IL-15 modulated the CD25, GITR, OX40, and CD73-phenotypes of the thymus-egress-competent and periphery-seeding Treg-population. Interestingly, IL-2 and IL-15 also modulated the TCR-repertoire expressed by developing Treg. Upon transfer into Treg-less Foxp3sf mice, newly developed Treg from IL-2- (and to a much lesser extent IL-15-) deficient mice suppressed immunopathology less efficiently than wt Treg. Taken together, our results firmly establish important non-redundant quantitative and qualitative roles for IL-2 and, to a lesser extent, IL-15 in intrathymic Treg-development.


Subject(s)
Interleukin-2 , T-Lymphocytes, Regulatory , Animals , Cytokines , Forkhead Transcription Factors/genetics , Mice , Receptors, Antigen, T-Cell
14.
J Clin Invest ; 118(6): 2098-110, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497879

ABSTRACT

The mouse CD8alpha+ DC subset excels at cross-presentation of antigen, which can elicit robust CTL responses. A receptor allowing specific antigen targeting to this subset and its equivalent in humans would therefore be useful for the induction of antitumor CTLs. Here, we have characterized a C-type lectin of the NK cell receptor group that we named DC, NK lectin group receptor-1 (DNGR-1). DNGR-1 was found to be expressed in mice at high levels by CD8+ DCs and at low levels by plasmacytoid DCs but not by other hematopoietic cells. Human DNGR-1 was also restricted in expression to a small subset of blood DCs that bear similarities to mouse CD8alpha+ DCs. The selective expression pattern and observed endocytic activity of DNGR-1 suggested that it could be used for antigen targeting to DCs. Consistent with this notion, antigen epitopes covalently coupled to an antibody specific for mouse DNGR-1 were selectively cross-presented by CD8alpha+ DCs in vivo and, when given with adjuvants, induced potent CTL responses. When the antigens corresponded to tumor-expressed peptides, treatment with the antibody conjugate and adjuvant could prevent development or mediate eradication of B16 melanoma lung pseudometastases. We conclude that DNGR-1 is a novel, highly specific marker of mouse and human DC subsets that can be exploited for CTL cross-priming and tumor therapy.


Subject(s)
CD8 Antigens/biosynthesis , Dendritic Cells/cytology , Immunotherapy/methods , Lectins/metabolism , Melanoma/pathology , Melanoma/therapy , Neoplasms/therapy , Receptors, Immunologic/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Animals , Antigens/chemistry , Dendritic Cells/immunology , Humans , Lectins, C-Type , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Models, Biological , Neoplasm Metastasis , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Receptors, Mitogen
15.
Eur J Immunol ; 40(5): 1255-65, 2010 May.
Article in English | MEDLINE | ID: mdl-20333625

ABSTRACT

DC NK lectin group receptor-1 (DNGR-1, also known as CLEC9A) is a C-type lectin receptor expressed by mouse CD8alpha+ DC and by their putative equivalents in human. DNGR-1 senses necrosis and regulates CD8+ T-cell cross-priming to dead-cell-associated antigens. In addition, DNGR-1 is a target for selective in vivo delivery of antigens to DC and the induction of CD8+ T-cell and Ab responses. In this study, we evaluated whether DNGR-1 targeting can be additionally used to manipulate antigen-specific CD8+ T lymphocytes. Injection of small amounts of antigen-coupled anti-DNGR-1 mAb into mice promoted MHC class II antigen presentation selectively by CD8alpha+ DC. In the steady state, this was sufficient to induce proliferation of antigen-specific naïve CD4+ T cells and to drive their differentiation into Foxp3+ regulatory lymphocytes. Co-administration of adjuvants prevented this induction of tolerance and promoted immunity. Notably, distinct adjuvants allowed qualitative modulation of CD4+ T-cell behavior: poly I:C induced a strong IL-12-independent Th1 response, whereas curdlan led to the priming of Th17 cells. Thus, antigen targeting to DNGR-1 is a versatile approach for inducing functionally distinct CD4+ T-cell responses. Given the restricted pattern of expression of DNGR-1 across species, this strategy could prove useful for developing immunotherapy protocols in humans.


Subject(s)
Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/analysis , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , T-Lymphocyte Subsets/immunology , Adjuvants, Immunologic , Animals , Antibodies, Monoclonal/pharmacology , Antigen Presentation , Epitopes , Forkhead Transcription Factors/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Peptide Fragments/immunology , Poly I-C/pharmacology , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , beta-Glucans/pharmacology
16.
Diabetes ; 70(8): 1729-1737, 2021 08.
Article in English | MEDLINE | ID: mdl-34035042

ABSTRACT

Regulatory T lymphocytes expressing the forkhead/winged helix transcription factor Foxp3 (Treg) play a vital role in the protection of the organism from autoimmune disease and other immunopathologies. The antigen specificity of Treg plays an important role in their in vivo activity. We therefore assessed the diversity of the T-cell receptors (TCRs) for antigen expressed by Treg newly developed in the thymus of autoimmune type 1 diabetes-prone NOD mice and compared it to the control mouse strain C57BL/6. Our results demonstrate that use of the TCRα and TCRß variable (V) and joining (J) segments, length of the complementarity determining region (CDR) 3, and the diversity of the TCRα and TCRß chains are comparable between NOD and C57BL/6 mice. Genetic defects affecting the diversity of the TCR expressed by newly developed Treg therefore do not appear to be involved in the etiology of type 1 diabetes in the NOD mouse.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/pathology , Thymus Gland/pathology , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology
17.
Eur J Immunol ; 39(2): 507-13, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19130473

ABSTRACT

Dectin-1 is a pattern-recognition receptor recognizing beta-(1,3)-glucans found on fungal cell walls. Dectin-1 plays an important role in immunity to fungi by mediating phagocytic clearance of fungal particles and inducing transcription of innate response genes. We show here that the two processes are linked and that Dectin-1 signalling for inflammation is attenuated by phagocytosis. Blocking Dectin-1 ligand-dependent internalization using either actin polymerization or dynamin inhibitors, large non-phagocytosable beta-glucan particles or poorly phagocytic cells leads in all cases to enhanced and sustained activation of downstream signalling pathways and culminates in production of high levels of pro-inflammatory cytokines. These findings establish the importance of phagocytosis not only in the clearance of pathogens, but also in the modulation of pattern-recognition receptor signalling and strongly suggest that internalization is the first step to attenuation of Dectin-1-mediated pro-inflammatory responses.


Subject(s)
Cytokines/immunology , Inflammation/immunology , Membrane Proteins/immunology , Mitogen-Activated Protein Kinase Kinases/immunology , Nerve Tissue Proteins/immunology , Phagocytosis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Immunity, Innate , Lectins, C-Type , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Nerve Tissue Proteins/genetics , Signal Transduction , beta-Glucans/immunology
18.
Med Sci (Paris) ; 36(3): 253-260, 2020 Mar.
Article in French | MEDLINE | ID: mdl-32228844

ABSTRACT

Upon priming by dendritic cells, naïve CD4 T lymphocytes are exposed to distinct molecular environments depending on the nature of the pathological stimulus. In response, they mobilize different gene networks that establish lineage-specific developmental programs, and coordinate the acquisition of specific phenotype and functions. Accordingly, CD4 T cells are capable of differentiation into a large variety of functionally-distinct T helper (Th) cell subsets. In this review, we describe the molecular events that control CD4 T cell differentiation at the level of the chromatin. We insist on recent works that have highlighted the key role of H3K9me3-dependent epigenetic mechanisms in the regulation of T cell identity. Interestingly, these pathways shape and control the developmental programs at least in part through the regulation of endogenous retroviruses-derived sequences that have been exapted into cis-regulatory modules of Th genes.


TITLE: Les rétrovirus endogènes - Un rôle clé dans la programmation des lymphocytes T CD4. ABSTRACT: Les lymphocytes T CD4 jouent un rôle clé dans le maintien de l'intégrité de l'organisme contre les dangers endogènes et exogènes. Ces cellules représentent donc un espoir thérapeutique majeur dans de nombreuses situations physiopathologiques. Dans cette synthèse, nous discuterons des mécanismes moléculaires qui définissent l'identité et les fonctions de ces cellules en réponse aux signaux de l'environnement. Nous nous intéresserons plus particulièrement aux voies épigénétiques qui coordonnent leur différenciation et leur plasticité. Des données récentes de la littérature suggèrent qu'elles pourraient agir en régulant l'activité de séquences dérivées de rétrovirus endogènes qui auraient été cooptées en modules cis-régulateurs de gènes pour le bénéfice de l'hôte.


Subject(s)
Endogenous Retroviruses/physiology , Immune System Phenomena/physiology , Animals , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Dendritic Cells/immunology , Dendritic Cells/physiology , Endogenous Retroviruses/genetics , Endogenous Retroviruses/immunology , Epigenesis, Genetic/physiology , Humans , Lymphocyte Activation/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology
19.
Cell Rep ; 32(7): 108048, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814051

ABSTRACT

During thymic development and upon peripheral activation, T cells undergo extensive phenotypic and functional changes coordinated by lineage-specific developmental programs. To characterize the regulatory landscape controlling T cell identity, we perform a wide epigenomic and transcriptional analysis of mouse thymocytes and naive CD4 differentiated T helper cells. Our investigations reveal a dynamic putative enhancer landscape, and we could validate many of the enhancers using the high-throughput CapStarr sequencing (CapStarr-seq) approach. We find that genes using multiple promoters display increased enhancer usage, suggesting that apparent "enhancer redundancy" might relate to isoform selection. Furthermore, we can show that two Runx3 promoters display long-range interactions with specific enhancers. Finally, our analyses suggest a novel function for the PRC2 complex in the control of alternative promoter usage. Altogether, our study has allowed for the mapping of an exhaustive set of active enhancers and provides new insights into their function and that of PRC2 in controlling promoter choice during T cell differentiation.


Subject(s)
Polycomb-Group Proteins/genetics , T-Lymphocytes/metabolism , Animals , Cell Differentiation , Male , Mice
20.
Front Immunol ; 9: 2555, 2018.
Article in English | MEDLINE | ID: mdl-30455700

ABSTRACT

While it is now acknowledged that CD4+ T cells expressing CD25 and Foxp3 (Treg cells) regulate immune responses and, consequently, influence the pathogenesis of infectious diseases, the regulatory response mediated by Treg cells upon infection by Trypanosoma cruzi was still poorly characterized. In order to understand the role of Treg cells during infection by this protozoan parasite, we determined in time and space the magnitude of the regulatory response and the phenotypic, functional and transcriptional features of the Treg cell population in infected mice. Contrary to the accumulation of Treg cells reported in most chronic infections in mice and humans, experimental T. cruzi infection was characterized by sustained numbers but decreased relative frequency of Treg cells. The reduction in Treg cell frequency resulted from a massive accumulation of effector immune cells, and inversely correlated with the magnitude of the effector immune response as well as with emergence of acute immunopathology. In order to understand the causes underlying the marked reduction in Treg cell frequency, we evaluated the dynamics of the Treg cell population and found a low proliferation rate and limited accrual of peripheral Treg cells during infection. We also observed that Treg cells became activated and acquired a phenotypic and transcriptional profile consistent with suppression of type 1 inflammatory responses. To assess the biological relevance of the relative reduction in Treg cells frequency observed during T. cruzi infection, we transferred in vitro differentiated Treg cells at early moments, when the deregulation of the ratio between regulatory and conventional T cells becomes significant. Intravenous injection of Treg cells dampened parasite-specific CD8+ T cell immunity and affected parasite control in blood and tissues. Altogether, our results show that limited Treg cell response during the acute phase of T. cruzi infection enables the emergence of protective anti-parasite CD8+ T cell immunity and critically influences host resistance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , T-Lymphocytes, Regulatory/immunology , Trypanosoma cruzi/immunology , Adoptive Transfer , Animals , Cell Proliferation , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL