scholarly journals Differential controls of MAIT cell effector polarization by mTORC1/mTORC2 via integrating cytokine and costimulatory signals

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Huishan Tao ◽  
Yun Pan ◽  
Shuai Chu ◽  
Lei Li ◽  
Jinhai Xie ◽  
...  

AbstractMucosal-associated invariant T (MAIT) cells have important functions in immune responses against pathogens and in diseases, but mechanisms controlling MAIT cell development and effector lineage differentiation remain unclear. Here, we report that IL-2/IL-15 receptor β chain and inducible costimulatory (ICOS) not only serve as lineage-specific markers for IFN-γ-producing MAIT1 and IL-17A-producing MAIT17 cells, but are also important for their differentiation, respectively. Both IL-2 and IL-15 induce mTOR activation, T-bet upregulation, and subsequent MAIT cell, especially MAIT1 cell, expansion. By contrast, IL-1β induces more MAIT17 than MAIT1 cells, while IL-23 alone promotes MAIT17 cell proliferation and survival, but synergizes with IL-1β to induce strong MAIT17 cell expansion in an mTOR-dependent manner. Moreover, mTOR is dispensable for early MAIT cell development, yet pivotal for MAIT cell effector differentiation. Our results thus show that mTORC2 integrates signals from ICOS and IL-1βR/IL-23R to exert a crucial role for MAIT17 differentiation, while the IL-2/IL-15R-mTORC1-T-bet axis ensures MAIT1 differentiation.

2017 ◽  
Author(s):  
Huimeng Wang ◽  
Criselle D’Souza ◽  
Xin Yi Lim ◽  
Lyudmila Kostenko ◽  
Troi J Pediongco ◽  
...  

AbstractMucosal associated invariant T (MAIT) cells recognize conserved microbial metabolites from riboflavin synthesis. Striking evolutionary conservation and pulmonary abundance implicate them in antibacterial host defense, yet their roles in protection against clinically significant pathogens are unknown. Murine Legionella infection induced MR1-dependent MAIT cell activation and rapid pulmonary accumulation of MAIT cells associated with immune protection detectable in fully immunocompetent host animals. MAIT cell protection was more evident in mice lacking CD4+ cells, whilst profoundly immunodeficient RAG2−/−γC−/− mice were substantially rescued from uniformly lethal Legionella infection by adoptively-transferred MAIT cells. This protection was dependent on MR1, IFN-γ and GM-CSF, but not IL-17, TNF-α or perforin. Protection was enhanced and observed earlier post-infection in mice that were Ag-primed to boost MAIT cells before infection. Our findings define a significant role for MAIT cells in protection against a major human pathogen and indicate a potential role for vaccination to enhance MAIT cell immunity.


Author(s):  
Tingting Liu ◽  
Jie Wang ◽  
Kalpana Subedi ◽  
Qijun Yi ◽  
Li Zhou ◽  
...  

Mucosal-associated invariant T (MAIT) cells are innate-like T cells that develop in the thymus through three maturation stages to acquire effector function and differentiate into MAIT1 (T-bet+) and MAIT17 (RORγt+) subsets. Upon activation, MAIT cells release IFN-γ and IL-17, which modulate a broad spectrum of diseases. Recent studies indicate defective MAIT cell development in microRNA deficient mice, however, few individual miRNAs have been identified to regulate MAIT cells. MicroRNA-155 (miR-155) is a key regulator of numerous cellular processes that affect some immune cell development, but its role in MAIT cell development remains unclear. To address whether miR-155 is required for MAIT cell development, we performed gain-of-function and loss-of-function studies. We first generated a CD4Cre.miR-155 knock-in mouse model, in which miR-155 is over-expressed in the T cell lineage. We found that overexpression of miR-155 significantly reduced numbers and frequencies of MAIT cells in all immune organs and lungs and blocked thymic MAIT cell maturation through downregulating PLZF expression. Strikingly, upregulated miR-155 promoted MAIT1 differentiation and blocked MAIT17 differentiation, and timely inducible expression of miR-155 functionally inhibited peripheral MAIT cells secreting IL-17. miR-155 overexpression also increased CD4–CD8+ subset and decreased CD4–CD8– subset of MAIT cells. We further analyzed MAIT cells in conventional miR-155 knockout mice and found that lack of miR-155 also promoted MAIT1 differentiation and blocked MAIT17 differentiation but without alteration of their overall frequency, maturation and function. Overall, our results indicate that adequate miR-155 expression is required for normal MAIT1 and MAIT17 cell development and function.


Author(s):  
Héloïse Flament ◽  
Matthieu Rouland ◽  
Lucie Beaudoin ◽  
Amine Toubal ◽  
Léo Bertrand ◽  
...  

Immune system dysfunction is paramount in Coronavirus disease 2019 (COVID-19) severity and fatality rate. Mucosal-Associated Invariant T (MAIT) cells are innate-like T cells involved in mucosal immunity and protection against viral infections. Here, we studied the immune cell landscape, with emphasis on MAIT cells, in a cohort of 182 patients including patients at various stages of disease activity. A profound decrease of MAIT cell counts in blood of critically ill patients was observed. These cells showed a strongly activated and cytotoxic phenotype that positively correlated with circulating pro-inflammatory cytokines, notably IL-18. MAIT cell alterations markedly correlated with disease severity and patient mortality. SARS-CoV-2-infected macrophages activated MAIT cells in a cytokine-dependent manner involving an IFNα-dependent early phase and an IL-18-induced later phase. Therefore, altered MAIT cell phenotypes represent valuable biomarkers of disease severity and their therapeutic manipulation might prevent the inflammatory phase involved in COVID-19 aggravation.


2019 ◽  
Author(s):  
Darshana Kadekar ◽  
Rasmus Agerholm ◽  
John Rizk ◽  
Heidi Neubauer ◽  
Tobias Suske ◽  
...  

SummaryInterleukin(IL)-17-producing RORγt+γδ T (γδT17) cells develop in the embryonic thymus and participate in type 3 immune responses. Herein we show that γδT17 cells rapidly proliferate within neonatal lymph nodes and gut, where upon entry they uniquely upregulate Tbet and co-express IL-17, IL-22 and interferon(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 homologue had a dominant role over STAT5B in promoting γδT17 cell expansion and downregulating gut-associated Tbet. In contrast, STAT5B preferentially expanded IFNγ-producing γδ populations. Importantly, mice lacking γδT17 cells due to STAT5 deficiency displayed a profound resistance to experimental autoimmune encephalomyelitis. Our data identify for the first time STAT5 as a key molecular checkpoint allowing γδT17 cells to pass through a critical neonatal developmental window to acquire tissue-specific characteristics essential for infection and autoimmunity.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 786-786
Author(s):  
Yuting Tang ◽  
Xiaomei Yan ◽  
Rui Huang ◽  
Yoshihiro Hayashi ◽  
Yue Zhang ◽  
...  

Abstract Natural killer (NK) cells are the major component of innate immunity with both cytotoxicity and cytokine producing effector functions. NK cells also regulate the interplay between innate immunity and adaptive immunity by secreting certain cytokines. Extrinsic regulators of NK cell development and function, including diverse ligands of NK cell receptors and cytokines from the microenvironment, have been extensively studied. However, intrinsic regulators for NK cell biology are still less understood. In our previous study on aggressive NK cell leukemia (ANKL), genomics and transcriptomics analyses indicated that c-MYC was universally upregulated and responsible for the proliferation and survival in ANKL cells (Manuscript in revision). Furthermore, STAT5, as a transcriptional regulator of c-MYC, was found to be essential in the survival and development of NK cells (Eckelhart et al., Blood 2011). In this regard, we want to understand the physiological and oncological roles of c-MYC in NK cells. To achieve our goal, we made two mouse models including c-Myc loss-of-function (LOF) and c-Myc gain-of-function (GOF) in NK cells. Ncr1Cre knock-in mice, in which Cre recombinase was inserted into the Nkp46 locus, was used. We crossed the c-Mycf/f mice with Ncr1Cre mice to generate the NK cell specific c-Myc LOF model c-MycΔ/Δ/Ncr1Cre. To generate the c-Myc GOF model, we crossed the Tg(tetO-MYC) mice with Ncr1Cre and Rosa26-Loxp-Stop-Loxp(LSL)-rtTA-GFP mice to get the Tg(tetO-MYC)/ Ncr1Cre/LSL-rtTA-GFP (iMYC) mice, in which c-Myc expression can be induced in a doxycycline dependent manner in NK cells. c-MycΔ/Δ/Ncr1Cre mice were analyzed between 6 to 14-weeks old. iMYC mice were induced by doxycycline from 6-weeks old for over 2 months and then analyzed. Wild type littermates were used as controls. In both models, mice were born normally and showed no obvious difference in growth compared to their littermates. In c-MycΔ/Δ/Ncr1Cre mice, a significant reduction of NK1.1+/DX5+ NK cell percentages in peripheral blood (3.6 ± 0.4% vs. 0.5 ± 0.1%, P < 0.0001, N=7) and spleen (2.4 ± 0.5% vs. 0.7 ± 0.1%, P < 0.01, N=6) was detected. In addition, the percentage of CD11b+ mature NK cells in the NK1.1+/DX5+ population was also reduced. In bone marrow (BM), although the total percentage of NK1.1+/DX5+ NK cells did not change, an obvious block of NK cell development was seen, as the majority of NK1.1+/DX5+ cells in BM were CD27+/CD11b- cells, which represent an immature pattern. To assess whether the NK cell proliferation is altered in this model, we performed BrdU labeling assays and found that BrdU incorporation rates decreased dramatically both in peripheral NK cells and BM NK progenitors. Functionally, we measured the IFN-γ secretion of splenic NK cells after PMA/Ionomycin stimulation. We found that the percentage of IFN-γ positive NK cells decreased significantly in c-MycΔ/Δ/Ncr1Cre mice (77.1 ± 7.0% vs. 53.8 ± 1.0%, N=3). Consistent with these data, the tumor surveillance was also severely impaired in this LOF model, as the number of lung metastatic sites significantly increased compared to the control mice in a B16F10 transplantation assay. In contrast to the LOF model, in our GOF model, the NK1.1+/DX5+ NK cell number in peripheral blood increased (3.1 ± 0.2% vs. 4.4 ± 0.5%, P < 0.05, N=7). Additionally, a small increase in the percentage of CD27-/CD11b+ population in NK1.1+/DX5+ cells was seen. Interestingly, however, the ability of IFN-γ secretion of splenic NK cells after PMA/Ionomycin stimulation was decreased in iMYC mice (72.8 ± 0.3% vs. 58.7 ± 1.1%, N=3), which showed the same alteration observed in c-MycΔ/Δ/Ncr1Cre mice. This result is probably not due to the impaired maturation, but rather it is the result of the higher percentage of CD27-/CD11b+ cells, which were considered terminally differentiated NK cells with lower cytotoxic functions. In summary, we found that c-Myc is essential for NK cell development, proliferation, and tumor surveillance. NK cell maturation and proliferation were impaired in the c-Myc LOF models and were boosted in the c-Myc GOF models. Our results also provide a mechanism basis for the potential application of targeting c-Myc in NK cells ex vivo or in vivo expansion, and NK-mediated immunotherapy. Future studies are needed to delineate the underlying mechanisms and explore the applications. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 86 (5) ◽  
Author(s):  
Eric Jesteadt ◽  
Irma Zhang ◽  
Huifeng Yu ◽  
Anda Meierovics ◽  
Wei-Jen Chua Yankelevich ◽  
...  

ABSTRACTMucosa-associated invariant T (MAIT) cells are a subset of innate T cells that express a semi-invariant Vα chain paired with limited Vβ chains. MAIT cells are activated by riboflavin metabolite derivatives presented by the nonpolymorphic major histocompatibility complex class I (MHC-I)-like molecule MR1. The precise mechanisms required to activate MAIT cells are an area of intense interest. Here we used two closely related intracellular pathogens with distinct inflammasome activation phenotypes to probe the role of innate cytokines in MAIT cell activation. Using anin vitroassay containing transgenic murine MAIT cells, we show that macrophages infected withFrancisella novicida, a strong inflammasome activator, released high levels of interleukin-18 (IL-18) and stimulated high levels of MAIT cell gamma interferon (IFN-γ) through a partially MR1-independent pathway. In contrast, macrophages infected withFrancisella tularensislive vaccine strain (LVS), a weak inflammasome activator, generated little IL-18 and stimulated low MAIT cell IFN-γ through an MR1-dependent pathway. By manipulating the quantities of IL-18 in these cultures, we show that the IL-18 concentration is sufficient to influence the magnitude of MAIT cell IFN-γ production. Correspondingly, infected IL-18-deficient macrophages failed to induce substantial MAIT cell IFN-γ. In contrast, we found that MAIT cell IFN-γ production in the lungs of IL-18-deficient mice was not significantly different from that in WT mice duringF. tularensisLVS pulmonary infection. Overall, we demonstrate that while IL-18 is essential for the MAIT cell IFN-γ responsein vitro, it is not essential for MAIT cell IFN-γ production duringin vivoLVS pulmonary infection, suggesting that additional signals can drive MAIT cell IFN-γ productionin vivo.


2019 ◽  
Vol 4 (41) ◽  
pp. eaay6039 ◽  
Author(s):  
H.-F. Koay ◽  
S. Su ◽  
D. Amann-Zalcenstein ◽  
S. R. Daley ◽  
I. Comerford ◽  
...  

MR1-restricted mucosal-associated invariant T (MAIT) cells play a unique role in the immune system. These cells develop intrathymically through a three-stage process, but the events that regulate this are largely unknown. Here, using bulk and single-cell RNA sequencing–based transcriptomic analysis in mice and humans, we studied the changing transcriptional landscape that accompanies transition through each stage. Many transcripts were sharply modulated during MAIT cell development, including SLAM (signaling lymphocytic activation molecule) family members, chemokine receptors, and transcription factors. We also demonstrate that stage 3 “mature” MAIT cells comprise distinct subpopulations including newly arrived transitional stage 3 cells, interferon-γ–producing MAIT1 cells and interleukin-17–producing MAIT17 cells. Moreover, the validity and importance of several transcripts detected in this study are directly demonstrated using specific mutant mice. For example, MAIT cell intrathymic maturation was found to be halted in SLAM-associated protein (SAP)–deficient and CXCR6-deficient mouse models, providing clear evidence for their role in modulating MAIT cell development. These data underpin a model that maps the changing transcriptional landscape and identifies key factors that regulate the process of MAIT cell differentiation, with many parallels between mice and humans.


2021 ◽  
Vol 12 ◽  
Author(s):  
Lichen Ouyang ◽  
Mi Wu ◽  
Zhijun Shen ◽  
Xue Cheng ◽  
Wei Wang ◽  
...  

Community-acquired pneumonia (CAP) remains the significant infectious cause of morbidity and mortality worldwide. Although mucosal-associated invariant T cells (MAIT) play roles in the pathogenesis of children CAP and ICU-associated pneumonia, their roles in adult CAP are largely unexplored. In this study, we investigated the frequency, phenotype, and function of MAIT cells in peripheral blood and bronchoalveolar lavage fluid (BALF) of adult CAP patients. Our data indicate that MAIT-cell frequency is profoundly lower in the peripheral blood of CAP patients compared to that in healthy individuals. Furthermore, the circulatory MAIT cells express higher levels of CD69 and PD-1 compared to those in healthy individuals. In BALF of CAP patients, MAIT-cell frequency is higher and MAIT cells express higher levels of CD69 and PD-1 compared to their matched blood counterparts. Levels of IL-17A and IFN-γ are increased in BALF of CAP patients compared to those in BALF of patients with pulmonary small nodules. The IL-17A/IFN-γ ratio is significantly positively correlated with MAIT frequency in BALF of CAP patients, suggesting a pathogenic role of MAIT-17 cells in CAP. Of note, blood MAIT-cell frequency in CAP patients is strongly negatively correlated with high-sensitivity C-reactive protein (hsCRP) and neutrophil count percentage in blood. The ability of circulating MAIT cells in CAP patients to produce IFN-γ is significantly impaired compared to those in healthy individuals. In summary, our findings suggest the possible involvement of MAIT cells in the immunopathogenesis of adult CAP.


2014 ◽  
Vol 211 (8) ◽  
pp. 1601-1610 ◽  
Author(s):  
Marielle C. Gold ◽  
James E. McLaren ◽  
Joseph A. Reistetter ◽  
Sue Smyk-Pearson ◽  
Kristin Ladell ◽  
...  

Mucosal-associated invariant T (MAIT) cells express a semi-invariant T cell receptor (TCR) that detects microbial metabolites presented by the nonpolymorphic major histocompatibility complex (MHC)–like molecule MR1. The highly conserved nature of MR1 in conjunction with biased MAIT TCRα chain usage is widely thought to indicate limited ligand presentation and discrimination within a pattern-like recognition system. Here, we evaluated the TCR repertoire of MAIT cells responsive to three classes of microbes. Substantial diversity and heterogeneity were apparent across the functional MAIT cell repertoire as a whole, especially for TCRβ chain sequences. Moreover, different pathogen-specific responses were characterized by distinct TCR usage, both between and within individuals, suggesting that MAIT cell adaptation was a direct consequence of exposure to various exogenous MR1-restricted epitopes. In line with this interpretation, MAIT cell clones with distinct TCRs responded differentially to a riboflavin metabolite. These results suggest that MAIT cells can discriminate between pathogen-derived ligands in a clonotype-dependent manner, providing a basis for adaptive memory via recruitment of specific repertoires shaped by microbial exposure.


2021 ◽  
Vol 12 ◽  
Author(s):  
Rosângela Salerno-Gonçalves ◽  
Tasmia Rezwan ◽  
David Luo ◽  
Hervé Tettelin ◽  
Marcelo B. Sztein

Mucosal-associated invariant T (MAIT) cells are an innate-like population of T cells that display a TCR Vα7.2+ CD161+ phenotype and are restricted by the nonclassical MHC-related molecule 1 (MR1). Although B cells control MAIT cell development and function, little is known about the mechanisms underlying their interaction(s). Here, we report, for the first time, that during Salmonella enterica serovar Typhi (S. Typhi) infection, HLA-G expression on B cells downregulates IFN-γ production by MAIT cells. In contrast, blocking HLA-G expression on S. Typhi-infected B cells increases IFN-γ production by MAIT cells. After interacting with MAIT cells, kinetic studies show that B cells upregulate HLA-G expression and downregulate the inhibitory HLA-G receptor CD85j on MAIT cells resulting in their loss. These results provide a new role for HLA-G as a negative feedback loop by which B cells control MAIT cell responses to antigens.


Sign in / Sign up

Export Citation Format

Share Document