Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Nat Immunol ; 15(8): 749-757, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973821

ABSTRACT

Interleukin 15 (IL-15) controls both the homeostasis and the peripheral activation of natural killer (NK) cells. The molecular basis for this duality of action remains unknown. Here we found that the metabolic checkpoint kinase mTOR was activated and boosted bioenergetic metabolism after exposure of NK cells to high concentrations of IL-15, whereas low doses of IL-15 triggered only phosphorylation of the transcription factor STAT5. mTOR stimulated the growth and nutrient uptake of NK cells and positively fed back on the receptor for IL-15. This process was essential for sustaining NK cell proliferation during development and the acquisition of cytolytic potential during inflammation or viral infection. The mTORC1 inhibitor rapamycin inhibited NK cell cytotoxicity both in mice and humans; this probably contributes to the immunosuppressive activity of this drug in different clinical settings.


Subject(s)
Interleukin-15/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , TOR Serine-Threonine Kinases/immunology , Animals , Cell Proliferation , Cells, Cultured , Herpesviridae Infections/immunology , Humans , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Killer Cells, Natural/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Muromegalovirus/immunology , Orthomyxoviridae Infections/immunology , Poly I-C/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
2.
Eur J Immunol ; : e2350954, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38837415

ABSTRACT

Hexokinases (HKs) control the first step of glucose catabolism. A switch of expression from liver HK (glucokinase, GCK) to the tumor isoenzyme HK2 is observed in hepatocellular carcinoma progression. Our prior work revealed that HK isoenzyme switch in hepatocytes not only regulates hepatic metabolic functions but also modulates innate immunity and sensitivity to Natural Killer (NK) cell cytotoxicity. This study investigates the impact of HK2 expression and its mitochondrial binding on the resistance of human liver cancer cells to NK-cell-induced cytolysis. We have shown that HK2 expression induces resistance to NK cell cytotoxicity in a process requiring mitochondrial binding of HK2. Neither HK2 nor GCK expression affects target cells' ability to activate NK cells. In contrast, mitochondrial binding of HK2 reduces effector caspase 3/7 activity both at baseline and upon NK-cell activation. Furthermore, HK2 tethering to mitochondria enhances their resistance to cytochrome c release triggered by tBID. These findings indicate that HK2 mitochondrial binding in liver cancer cells is an intrinsic resistance factor to cytolysis and an escape mechanism from immune surveillance.

3.
J Immunol ; 210(9): 1209-1221, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36961448

ABSTRACT

Autosomal recessive PRKCD deficiency has previously been associated with the development of systemic lupus erythematosus in human patients, but the mechanisms underlying autoimmunity remain poorly understood. We introduced the Prkcd G510S mutation that we previously associated to a Mendelian cause of systemic lupus erythematosus in the mouse genome, using CRISPR-Cas9 gene editing. PrkcdG510S/G510S mice recapitulated the human phenotype and had reduced lifespan. We demonstrate that this phenotype is linked to a B cell-autonomous role of Prkcd. A detailed analysis of B cell activation in PrkcdG510S/G510S mice shows an upregulation of the PI3K/mTOR pathway after the engagement of the BCR in these cells, leading to lymphoproliferation. Treatment of mice with rapamycin, an mTORC1 inhibitor, significantly improves autoimmune symptoms, demonstrating in vivo the deleterious effect of mTOR pathway activation in PrkcdG510S/G510S mice. Additional defects in PrkcdG510S/G510S mice include a decrease in peripheral mature NK cells that might contribute to the known susceptibility to viral infections of patients with PRKCD mutations.


Subject(s)
Autoimmunity , Lupus Erythematosus, Systemic , Humans , Animals , Mice , TOR Serine-Threonine Kinases/metabolism , B-Lymphocytes , Cell Proliferation
4.
J Immunol ; 208(7): 1802-1812, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35288470

ABSTRACT

NK cell receptors allow NK cells to recognize targets such as tumor cells. Many of them are expressed on a subset of NK cells, independently of each other, which creates a vast diversity of receptor combinations. Whether these combinations influence NK cell antitumor responses is not well understood. We addressed this question in the C57BL/6 mouse model and analyzed the individual effector response of 444 mouse NK cell subsets, defined by combinations of 12 receptors, against tumor cell lines originating from different tissues and mouse strains. We found a wide range of reactivity among NK subsets, but the same hierarchy of responses was observed for the different tumor types, showing that the repertoire of NK cell receptors does not encode for different tumor specificities but for different intrinsic reactivities. The coexpression of CD27, NKG2A, and DNAM-1 identified subsets with relative cytotoxic specialization, whereas reciprocally, CD11b and KLRG1 defined the best IFN-γ producers. The expression of educating receptors Ly49C, Ly49I, and NKG2A was also strongly correlated with IFN-γ production, but this effect was suppressed by unengaged receptors Ly49A, Ly49F, and Ly49G2. Finally, IL-15 coordinated NK cell effector functions, but education and unbound inhibitory receptors retained some influence on their response. Collectively, these data refine our understanding of the mechanisms governing NK cell reactivity, which could help design new NK cell therapy protocols.


Subject(s)
Interferon-gamma , Killer Cells, Natural , Animals , Cell Line, Tumor , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Receptors, Natural Killer Cell/metabolism
5.
J Immunol ; 206(10): 2265-2270, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33931486

ABSTRACT

NK cells are cytotoxic lymphocytes displaying strong antimetastatic activity. Mouse models and in vitro studies suggest a prominent role of the mechanistic target of rapamycin (mTOR) kinase in the control of NK cell homeostasis and antitumor functions. However, mTOR inhibitors are used as chemotherapies in several cancer settings. The impact of such treatments on patients' NK cells is unknown. We thus performed immunophenotyping of circulating NK cells from metastatic breast cancer patients treated with the mTOR inhibitor everolimus over a three-month period. Everolimus treatment resulted in inhibition of mTORC1 activity in peripheral NK cells, whereas mTORC2 activity was preserved. NK cell homeostasis was profoundly altered with a contraction of the NK cell pool and an overall decrease in their maturation. Phenotype and function of the remaining NK cell population was less affected. This is, to our knowledge, the first in vivo characterization of the role of mTOR in human NK cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Everolimus/administration & dosage , Killer Cells, Natural/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/drug effects , Adult , Aged , Aged, 80 and over , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Female , Follow-Up Studies , France/epidemiology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Middle Aged , Neoplasm Metastasis , Prospective Studies , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
6.
Int J Mol Sci ; 24(16)2023 Aug 13.
Article in English | MEDLINE | ID: mdl-37628917

ABSTRACT

CD8+ T cells and Natural Killer (NK) cells are cytotoxic lymphocytes important in the response to intracellular pathogens and cancer. Their activity depends on the integration of a large set of intracellular and environmental cues, including antigenic signals, cytokine stimulation and nutrient availability. This integration is achieved by signaling hubs, such as the mechanistic target of rapamycin (mTOR). mTOR is a conserved protein kinase that controls cellular growth and metabolism in eukaryotic cells and, therefore, is essential for lymphocyte development and maturation. However, our current understanding of mTOR signaling comes mostly from studies performed in transformed cell lines, which constitute a poor model for comprehending metabolic pathway regulation. Therefore, it is only quite recently that the regulation of mTOR in primary cells has been assessed. Here, we review the signaling pathways leading to mTOR activation in CD8+ T and NK cells, focusing on activation by cytokines. We also discuss how this knowledge can contribute to immunotherapy development, particularly for cancer treatment.


Subject(s)
Signal Transduction , TOR Serine-Threonine Kinases , Killer Cells, Natural , CD8-Positive T-Lymphocytes , Cell Cycle , Cytokines
7.
J Immunol ; 200(2): 551-557, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29237776

ABSTRACT

The role of sphingosine-1 phosphate (S1P) in leukocyte trafficking has been well deciphered in mice but remains largely unaddressed in humans. In this study, we assessed the ex vivo response to S1P of primary human T cell subsets. We found that tonsil but not blood leukocytes were responsive to S1P gradients, suggesting that T cell responsiveness is regulated during their recirculation in vivo. Tonsil naive T cells were readily chemoattracted by S1P in an FTY720-sensitive, S1PR1-dependent manner. Surprisingly, S1P had the opposite effect on effector memory T cells, resident memory T cells, and recently activated T cells, inhibiting their spontaneous or chemokine-induced migration. This inhibition was also more pronounced for CD4 T cells than for CD8 T cell subsets, and was dependent on S1PR2, as shown using the S1PR2 antagonist JTE-013. S1PR1 was progressively downregulated during T cell differentiation whereas S1PR2 expression remained stable. Our results suggest that the ratio between S1PR1 and S1PR2 governs the migratory behavior of T cell subsets. They also challenge previous models of the role of S1P in lymphocyte recirculation and suggest that S1P promotes retention of memory T cell subsets in secondary lymphoid organs, via S1PR2.


Subject(s)
Chemotaxis, Leukocyte/immunology , Immunologic Memory , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , T-Lymphocytes/immunology , Chemokines/metabolism , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/immunology , Sphingosine-1-Phosphate Receptors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism
8.
Eur J Immunol ; 48(5): 738-750, 2018 05.
Article in English | MEDLINE | ID: mdl-29424438

ABSTRACT

T-bet and Eomes are T-box transcription factors that drive the differentiation and function of cytotoxic lymphocytes such as NK cells. Their DNA-binding domains are highly similar, suggesting redundant transcriptional activity. However, while these transcription factors have different patterns of expression, the phenotype of loss-of-function mouse models suggests that they play distinct roles in the development of NK cells and other innate lymphoid cells (ILCs). Recent technological advances using reporter mice and conditional knockouts were fundamental in defining the regulation and function of these factors at steady state and during pathological conditions such as various types of cancer or infection. Here, we review these recent developments, focusing on NK cells as prototypical cytotoxic lymphocytes and their development, and also discuss parallels between NK cells and T cells. We also examine the role of T-bet and Eomes in human NK cells and ILC1s. Considering divergent findings on mouse and human ILC1s, we propose that NK cells are defined by coexpression of T-bet and Eomes, while ILC1s express only one of these factors, either T-bet or Eomes, depending on the tissue or the species.


Subject(s)
Killer Cells, Natural/cytology , Lymphocyte Subsets/cytology , T-Box Domain Proteins/metabolism , Animals , Cell Differentiation , Communicable Diseases/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Mice , Neoplasms/immunology , T-Box Domain Proteins/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
9.
PLoS Genet ; 11(2): e1005020, 2015.
Article in English | MEDLINE | ID: mdl-25714103

ABSTRACT

The development and homeostasis of multicellular organisms relies on gene regulation within individual constituent cells. Gene regulatory circuits that increase the robustness of gene expression frequently incorporate microRNAs as post-transcriptional regulators. Computational approaches, synthetic gene circuits and observations in model organisms predict that the co-regulation of microRNAs and their target mRNAs can reduce cell-to-cell variability in the expression of target genes. However, whether microRNAs directly regulate variability of endogenous gene expression remains to be tested in mammalian cells. Here we use quantitative flow cytometry to show that microRNAs impact on cell-to-cell variability of protein expression in developing mouse thymocytes. We find two distinct mechanisms that control variation in the activation-induced expression of the microRNA target CD69. First, the expression of miR-17 and miR-20a, two members of the miR-17-92 cluster, is co-regulated with the target mRNA Cd69 to form an activation-induced incoherent feed-forward loop. Another microRNA, miR-181a, acts at least in part upstream of the target mRNA Cd69 to modulate cellular responses to activation. The ability of microRNAs to render gene expression more uniform across mammalian cell populations may be important for normal development and for disease.


Subject(s)
Cell Survival/genetics , MicroRNAs/genetics , Protein Biosynthesis/genetics , Thymocytes/metabolism , Animals , Cell Line, Tumor , Flow Cytometry , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Mice , RNA, Messenger/biosynthesis
10.
Clin Immunol ; 177: 12-17, 2017 04.
Article in English | MEDLINE | ID: mdl-26794911

ABSTRACT

Obesity is associated with increased cancer rates and higher susceptibility to infections. The adipose tissue of obese individuals is inflammatory and may negatively impact on innate and adaptive immunity in a systemic way. Here, we explored the phenotype and function of peripheral Natural Killer (NK) cells of patients in correlation with their body mass index (BMI). We found that high BMI was associated with an increased activation status of peripheral NK cells, as measured by surface levels of CD69 and levels of granzyme-B. However, these activated NK cells had an impaired capacity to degranulate or to produce cytokines/chemokines when exposed to tumor cell lines deficient in MHC-I expression or coated with antibodies. This suggests that chronic stimulation of NK cells during obesity may lead to their incapacity to respond normally and eliminate target cells, which could contribute to the greater susceptibility of obese individuals to develop cancers or infectious diseases.


Subject(s)
Killer Cells, Natural/immunology , Obesity/immunology , Adult , Aged , Female , Humans , Male , Middle Aged , Phenotype , Young Adult
11.
Eur J Immunol ; 46(8): 1902-11, 2016 08.
Article in English | MEDLINE | ID: mdl-27195970

ABSTRACT

MicroRNAs control developmental pathways and effector functions in immune cells. Previous studies have studied the role of microRNAs in natural killer (NK) cells. However, the mouse models of microRNA depletion used were nonNK-specific and only partially depleting, hampering the interpretation of the data obtained. To clarify the role of microRNAs in murine NK cells, we deleted the RNase III enzyme Dicer1 in NKp46-expressing cells. We observed a drastic decrease in several microRNAs specifically in NK cells. Furthermore, the overall size of the "NK-cell" pool was severely decreased, a phenotype associated with compromised survival. Moreover, performing a broad flow cytometry profiling, we show that Dicer1-deficient NK cells failed to complete their differentiation program. In particular, several integrins were inappropriately expressed in mature NK cells. These defects coincided with decreased response to IL-15, a cytokine responsible for "NK-cell" maturation and survival. In addition, Dicer1 deletion impaired key "NK-cell" functions: target cell killing and production of IFN-γ, leading to defective control of metastasis. Dicer1 deletion thus affects "NK-cell" biology in a cell intrinsic manner at several distinct stages.


Subject(s)
Antigens, Ly/metabolism , DEAD-box RNA Helicases/metabolism , Killer Cells, Natural/immunology , MicroRNAs/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Ribonuclease III/metabolism , Animals , Antigens, Ly/genetics , Cell Differentiation , Cytotoxicity, Immunologic , DEAD-box RNA Helicases/genetics , Female , Flow Cytometry , Hematopoiesis , Interferon-gamma/metabolism , Interleukin-15/metabolism , Lymphocyte Activation , Mice , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/genetics , Ribonuclease III/genetics , Signal Transduction
12.
Eur J Immunol ; 46(9): 2095-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27600673

ABSTRACT

Recent studies of immune populations in nonlymphoid organs have highlighted the great diversity of the innate lymphoid system. It has also become apparent that mouse and human innate lymphoid cells (ILCs) have distinct phenotypes and properties. In this issue of the European Journal of Immunology, Harmon et al. [Eur. J. Immunol. 2016. 46: 2111-2120] characterized human hepatic NK-cell subsets. The authors report that hepatic CD56(bright) NK cells resemble mouse liver ILC1s in that they express CXCR6 and have an immature phenotype. However, unlike mouse ILC1s, they express high levels of Eomes and low levels of T-bet, and upon stimulation with tumor cells, secrete low amounts of cytokines. These unexpected findings further support the differences between human and mouse immune populations and prompt the study of the role of hepatic ILC subsets in immune responses.


Subject(s)
Immunity, Innate/immunology , Killer Cells, Natural/immunology , Animals , Cytokines , Hepatocytes , Humans , Liver/immunology , Mice
13.
Nucleic Acids Res ; 42(17): e135, 2014.
Article in English | MEDLINE | ID: mdl-25081207

ABSTRACT

microRNAs (miRNAs) are a class of ∼22nt non-coding RNAs that potentially regulate over 60% of human protein-coding genes. miRNA activity is highly specific, differing between cell types, developmental stages and environmental conditions, so the identification of active miRNAs in a given sample is of great interest. Here we present a novel computational approach for analyzing both mRNA sequence and gene expression data, called MixMir. Our method corrects for 3' UTR background sequence similarity between transcripts, which is known to correlate with mRNA transcript abundance. We demonstrate that after accounting for kmer sequence similarities in 3' UTRs, a statistical linear model based on motif presence/absence can effectively discover active miRNAs in a sample. MixMir utilizes fast software implementations for solving mixed linear models, which are widely used in genome-wide association studies (GWASs). Essentially we use 3' UTR sequence similarity in place of population cryptic relatedness in the GWAS problem. Compared to similar methods such as miReduce, Sylamer and cWords, we found that MixMir performed better at discovering true miRNA motifs in three mouse Dicer-knockout experiments from different tissues, two of which were collected by our group. We confirmed these results on protein and mRNA expression data obtained from miRNA transfection experiments in human cell lines. MixMir can be freely downloaded from https://github.com/ldiao/MixMir.


Subject(s)
3' Untranslated Regions , Gene Expression Profiling/methods , MicroRNAs/metabolism , Sequence Analysis, RNA/methods , Adrenal Cortex/metabolism , Algorithms , Animals , DEAD-box RNA Helicases/genetics , Embryonic Stem Cells/metabolism , Linear Models , Mice , Mice, Knockout , Nucleotide Motifs , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribonuclease III/genetics
14.
J Immunol ; 189(7): 3480-9, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22942430

ABSTRACT

IL-4 is one of the main cytokines produced during Th2-inducing pathologies. This cytokine has been shown to affect a number of immune processes such as Th differentiation and innate immune responses. However, the impact of IL-4 on CD8 T cell responses remains unclear. In this study, we analyzed the effects of IL-4 on global gene expression profiles of Ag-induced memory CD8 T cells in the mouse. Gene ontology analysis of this signature revealed that IL-4 regulated most importantly genes associated with immune responses. Moreover, this IL-4 signature overlapped with the set of genes preferentially expressed by memory CD8 T cells over naive CD8 T cells. In particular, IL-4 downregulated in vitro and in vivo in a STAT6-dependent manner the memory-specific expression of NKG2D, thereby increasing the activation threshold of memory CD8 T cells. Furthermore, IL-4 impaired activation of memory cells as well as their differentiation into effector cells. This phenomenon could have an important clinical relevance as patients affected by Th2 pathologies such as parasitic infections or atopic dermatitis often suffer from viral-induced complications possibly linked to inefficient CD8 T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Down-Regulation/immunology , Immunologic Memory , Interleukin-4/physiology , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Animals , Cells, Cultured , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/metabolism , Down-Regulation/genetics , Humans , Immunity, Innate/genetics , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/genetics , STAT6 Transcription Factor/physiology
16.
J Immunol ; 183(4): 2506-12, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19625646

ABSTRACT

Invariant NK T (iNKT) cells are a separate lineage of T lymphocytes with innate effector functions. They express an invariant TCR specific for lipids presented by CD1d and their development and effector differentiation rely on a unique gene expression program. We asked whether this program includes microRNAs, small noncoding RNAs that regulate gene expression posttranscriptionally and play a key role in the control of cellular differentiation programs. To this aim, we investigated iNKT cell development in mice in which Dicer, the RNase III enzyme that generates functional microRNAs, is deleted in cortical thymocytes. We find that Dicer deletion results in a substantial reduction of iNKT cells in thymus and their disappearance from the periphery, unlike mainstream T cells. Without Dicer, iNKT cells do not complete their innate effector differentiation and display a defective homeostasis due to increased cell death. Differentiation and homeostasis of iNKT cells require Dicer in a cell-autonomous fashion. Furthermore, we identify a miRNA profile specific for iNKT cells, which exhibits features of activated/effector T lymphocytes, consistent with the idea that iNKT cells undergo agonist thymic selection. Together, these results define a critical role of the Dicer-dependent miRNA pathway in the physiology of iNKT cells.


Subject(s)
Cell Differentiation/immunology , DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Enzymologic/immunology , MicroRNAs/genetics , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Animals , Cell Differentiation/genetics , Gene Expression Profiling , Growth Inhibitors/genetics , Lymphopenia/enzymology , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , Natural Killer T-Cells/cytology , Ribonuclease III , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/enzymology , Thymus Gland/immunology
17.
J Immunol ; 182(6): 3846-54, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265164

ABSTRACT

Most memory CD8 T cell subsets that have been hitherto defined are generated in response to infectious pathogens. In this study, we have characterized the CD8 T cells that survive priming conditions, devoid of pathogen-derived danger signals. In both a TCR-transgenic model and a model of contact hypersensitivity, we show that the priming of naive CD8 T cells under sterile inflammatory conditions generates memory. The corresponding memory CD8 T cells can be identified by their intermediate expression levels of CD44 and CD122. We also show that CD44/122(int) memory CD8 T cells spontaneously develop in wild type mice and that they display intermediate levels of several other memory traits including functional (IFN-gamma secretion capacity, CCL5 messenger stores), phenotypic, and molecular (T-bet and eomesodermin expression levels) features. We finally show that they correspond to an early differentiation stage and can further differentiate in CD44/122(high) memory T cells. Altogether, our results identify a new memory CD8 T cell subset that is generated under sterile inflammatory conditions and involved in the recall contact hypersensitivity reactions that are responsible for allergic contact dermatitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Hyaluronan Receptors/physiology , Immunologic Memory/genetics , Inflammation Mediators/physiology , Interleukin-2 Receptor beta Subunit/physiology , Lymphocyte Activation/immunology , Animals , Biomarkers/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Hyaluronan Receptors/biosynthesis , Inflammation Mediators/metabolism , Interleukin-2 Receptor beta Subunit/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
Elife ; 102021 01 28.
Article in English | MEDLINE | ID: mdl-33507150

ABSTRACT

Antiviral effectors such as natural killer (NK) cells have impaired functions in chronic hepatitis B (CHB) patients. The molecular mechanism responsible for this dysfunction remains poorly characterised. We show that decreased cytokine production capacity of peripheral NK cells from CHB patients was associated with reduced expression of NKp30 and CD16, and defective mTOR pathway activity. Transcriptome analysis of patients NK cells revealed an enrichment for transcripts expressed in exhausted T cells suggesting that NK cell dysfunction and T cell exhaustion employ common mechanisms. In particular, the transcription factor TOX and several of its targets were over-expressed in NK cells of CHB patients. This signature was predicted to be dependent on the calcium-associated transcription factor NFAT. Stimulation of the calcium-dependent pathway recapitulated features of NK cells from CHB patients. Thus, deregulated calcium signalling could be a central event in both T cell exhaustion and NK cell dysfunction occurring during chronic infections.


Subject(s)
Antiviral Restriction Factors/immunology , Hepatitis B, Chronic/immunology , Killer Cells, Natural/immunology , Persistent Infection/immunology , T-Lymphocytes/immunology , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
19.
Cell Mol Immunol ; 18(9): 2140-2152, 2021 09.
Article in English | MEDLINE | ID: mdl-32398809

ABSTRACT

T cell development proceeds under the influence of a network of transcription factors (TFs). The precise role of Zeb1, a member of this network, remains unclear. Here, we report that Zeb1 expression is induced early during T cell development in CD4-CD8- double-negative (DN) stage 2 (DN2). Zeb1 expression was further increased in the CD4+CD8+ double-positive (DP) stage before decreasing in more mature T cell subsets. We performed an exhaustive characterization of T cells in Cellophane mice that bear Zeb1 hypomorphic mutations. The Zeb1 mutation profoundly affected all thymic subsets, especially DN2 and DP cells. Zeb1 promoted the survival and proliferation of both cell populations in a cell-intrinsic manner. In the periphery of Cellophane mice, the number of conventional T cells was near normal, but invariant NKT cells, NK1.1+ γδ T cells and Ly49+ CD8 T cells were virtually absent. This suggested that Zeb1 regulates the development of unconventional T cell types from DP progenitors. A transcriptomic analysis of WT and Cellophane DP cells revealed that Zeb1 regulated the expression of multiple genes involved in the cell cycle and TCR signaling, which possibly occurred in cooperation with Tcf1 and Heb. Indeed, Cellophane DP cells displayed stronger signaling than WT DP cells upon TCR engagement in terms of the calcium response, phosphorylation events, and expression of early genes. Thus, Zeb1 is a key regulator of the cell cycle and TCR signaling during thymic T cell development. We propose that thymocyte selection is perturbed in Zeb1-mutated mice in a way that does not allow the survival of unconventional T cell subsets.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocyte Subsets , Animals , Cell Differentiation , Cell Proliferation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction/genetics , Thymus Gland
20.
Nat Commun ; 12(1): 5446, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521844

ABSTRACT

EOMES and T-BET are related T-box transcription factors that control natural killer (NK) cell development. Here we demonstrate that EOMES and T-BET regulate largely distinct gene sets during this process. EOMES is dominantly expressed in immature NK cells and drives early lineage specification by inducing hallmark receptors and functions. By contrast, T-BET is dominant in mature NK cells, where it induces responsiveness to IL-12 and represses the cell cycle, likely through transcriptional repressors. Regardless, many genes with distinct functions are co-regulated by the two transcription factors. By generating two gene-modified mice facilitating chromatin immunoprecipitation of endogenous EOMES and T-BET, we show a strong overlap in their DNA binding targets, as well as extensive epigenetic changes during NK cell differentiation. Our data thus suggest that EOMES and T-BET may distinctly govern, via differential expression and co-factors recruitment, NK cell maturation by inserting partially overlapping epigenetic regulations.


Subject(s)
Cell Cycle/genetics , Cell Lineage/genetics , Killer Cells, Natural/immunology , T-Box Domain Proteins/genetics , Animals , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Cycle/drug effects , Cell Cycle/immunology , Cell Differentiation , Cell Lineage/drug effects , Cell Lineage/immunology , Epigenesis, Genetic/immunology , Interleukin-12/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Protein Binding , Spleen/cytology , Spleen/immunology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/immunology , Transcription, Genetic , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL