Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
2.
Proc Natl Acad Sci U S A ; 121(17): e2318420121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38621136

ABSTRACT

In response to an immune challenge, naive T cells undergo a transition from a quiescent to an activated state acquiring the effector function. Concurrently, these T cells reprogram cellular metabolism, which is regulated by iron. We and others have shown that iron homeostasis controls proliferation and mitochondrial function, but the underlying mechanisms are poorly understood. Given that iron derived from heme makes up a large portion of the cellular iron pool, we investigated iron homeostasis in T cells using mice with a T cell-specific deletion of the heme exporter, FLVCR1 [referred to as knockout (KO)]. Our finding revealed that maintaining heme and iron homeostasis is essential to keep naive T cells in a quiescent state. KO naive CD4 T cells exhibited an iron-overloaded phenotype, with increased spontaneous proliferation and hyperactive mitochondria. This was evidenced by reduced IL-7R and IL-15R levels but increased CD5 and Nur77 expression. Upon activation, however, KO CD4 T cells have defects in proliferation, IL-2 production, and mitochondrial functions. Iron-overloaded CD4 T cells failed to induce mitochondrial iron and exhibited more fragmented mitochondria after activation, making them susceptible to ferroptosis. Iron overload also led to inefficient glycolysis and glutaminolysis but heightened activity in the hexosamine biosynthetic pathway. Overall, these findings highlight the essential role of iron in controlling mitochondrial function and cellular metabolism in naive CD4 T cells, critical for maintaining their quiescent state.


Subject(s)
CD4-Positive T-Lymphocytes , Iron , Mice , Animals , Iron/metabolism , Mitochondria/metabolism , Signal Transduction , Heme/metabolism
3.
Immunohorizons ; 7(3): 235-242, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36951874

ABSTRACT

The E3 ubiquitin ligase cullin 3 (Cul3) is critical for invariant NKT (iNKT) cell development, as iNKT cells lacking Cul3 accumulate in the immature developmental stages. However, the mechanisms by which Cul3 mediates iNKT cell development remain unknown. In this study, we investigated the role of Cul3 in both immature and mature thymic iNKT cells using a mouse model with a T cell-specific deletion of Cul3. We found that mature iNKT cells lacking Cul3 proliferated and died more than wild-type cells did. These cells also displayed increased glucose metabolism and autophagy. Interestingly, we found that tight regulation of iron homeostasis is critical for iNKT cell development. Without Cul3, mature iNKT cells harbored higher levels of cytosolic iron, a phenotype associated with increased cell death. Taken together, our data suggest that Cul3 promotes iNKT cell development partially through intracellular iron homeostasis.


Subject(s)
Natural Killer T-Cells , Animals , Mice , Cell Differentiation/genetics , Natural Killer T-Cells/metabolism , Mice, Knockout , Cullin Proteins/genetics , Cullin Proteins/metabolism , Homeostasis
4.
Cell Rep ; 41(4): 111516, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36288696

ABSTRACT

Natural killer T (NKT) cells operate distinctly different metabolic programming from CD4 T cells, including a strict requirement for glutamine to regulate cell homeostasis. However, the underlying mechanisms remain unknown. Here, we report that at a steady state, NKT cells have higher glutamine levels than CD4 T cells and that NKT cells increase glutaminolysis on activation. Activated NKT cells use glutamine to fuel the tricarboxylic acid cycle and glutathione synthesis. In addition, glutamine-derived nitrogen enables protein glycosylation via the hexosamine biosynthesis pathway (HBP). Each of these branches of glutamine metabolism seems to be critical for NKT cell homeostasis and mitochondrial functions. Glutaminolysis and HBP differentially regulate interleukin-4 (IL-4) and interferon γ (IFNγ) production. Glutamine metabolism appears to be controlled by AMP-activated protein kinase (AMPK)-mammalian target of rapamycin complex 1 (mTORC1) signaling. These findings highlight a distinct metabolic requirement of NKT cells compared with CD4 T cells, which may have therapeutic implications in the treatment of certain nutrient-restricted diseases.


Subject(s)
Natural Killer T-Cells , Natural Killer T-Cells/metabolism , Interleukin-4/metabolism , Glutamine/metabolism , AMP-Activated Protein Kinases/metabolism , Interferon-gamma/metabolism , Homeostasis , Hexosamines/metabolism , Phenotype , TOR Serine-Threonine Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Nitrogen/metabolism , Glutathione/metabolism
5.
Immunometabolism ; 3(2)2021.
Article in English | MEDLINE | ID: mdl-33717605

ABSTRACT

Cellular metabolism is critical for generating energy and macromolecules for cell growth and survival. In recent years, the importance of metabolism in mediating T cell differentiation, proliferation, and function has been a hot topic of investigation. However, very little is known about metabolic regulation in invariant natural killer T (iNKT) cells. In this viewpoint, we will discuss what is currently known about immunometabolism in iNKT cells and how these findings relate to CD4 T cells.

6.
J Immunol ; 204(7): 1708-1713, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32122995

ABSTRACT

Iron has long been established as a critical mediator of T cell development and proliferation. However, the mechanisms by which iron controls CD4 T cell activation and expansion remain poorly understood. In this study, we show that stimulation of CD4 T cells from C57BL/6 mice not only decreases total and labile iron levels but also leads to changes in the expression of iron homeostatic machinery. Additionally, restraining iron availability in vitro severely inhibited CD4 T cell proliferation and cell cycle progression. Although modulating cellular iron levels increased IL-2 production by activated T lymphocytes, CD25 expression and pSTAT5 levels were decreased, indicating that iron is necessary for IL-2R-mediated signaling. We also found that iron deprivation during T cell stimulation negatively impacts mitochondrial function, which can be reversed by iron supplementation. In all, we show that iron contributes to activation-induced T cell expansion by positively regulating IL-2R signaling and mitochondrial function.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Iron/immunology , Mitochondria/immunology , Receptors, Interleukin-2/immunology , Animals , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
7.
PLoS Pathog ; 15(10): e1007903, 2019 10.
Article in English | MEDLINE | ID: mdl-31584995

ABSTRACT

HIV-1 gene expression is regulated by host and viral factors that interact with viral motifs and is influenced by proviral integration sites. Here, expression variation among integrants was followed for hundreds of individual proviral clones within polyclonal populations throughout successive rounds of virus and cultured cell replication, with limited findings using CD4+ cells from donor blood consistent with observations in immortalized cells. Tracking clonal behavior by proviral "zip codes" indicated that mutational inactivation during reverse transcription was rare, while clonal expansion and proviral expression states varied widely. By sorting for provirus expression using a GFP reporter in the nef open reading frame, distinct clone-specific variation in on/off proportions were observed that spanned three orders of magnitude. Tracking GFP phenotypes over time revealed that as cells divided, their progeny alternated between HIV transcriptional activity and non-activity. Despite these phenotypic oscillations, the overall GFP+ population within each clone was remarkably stable, with clones maintaining clone-specific equilibrium mixtures of GFP+ and GFP- cells. Integration sites were analyzed for correlations between genomic features and the epigenetic phenomena described here. Integrants inserted in the sense orientation of genes were more frequently found to be GFP negative than those in the antisense orientation, and clones with high GFP+ proportions were more distal to repressive H3K9me3 peaks than low GFP+ clones. Clones with low frequencies of GFP positivity appeared to expand more rapidly than clones for which most cells were GFP+, even though the tested proviruses were Vpr-. Thus, much of the increase in the GFP- population in these polyclonal pools over time reflected differential clonal expansion. Together, these results underscore the temporal and quantitative variability in HIV-1 gene expression among proviral clones that are conferred in the absence of metabolic or cell-type dependent variability, and shed light on cell-intrinsic layers of regulation that affect HIV-1 population dynamics.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , Proviruses/genetics , Virus Integration/genetics , Virus Replication , CD4-Positive T-Lymphocytes/metabolism , HIV Infections/genetics , High-Throughput Screening Assays , Humans , Jurkat Cells , Transduction, Genetic
8.
Cell Rep ; 27(3): 699-707.e4, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995469

ABSTRACT

Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) proteins work in concert to regulate the levels of reactive oxygen species (ROS). The Keap1-Nrf2 antioxidant system also participates in T cell differentiation and inflammation, but its role in innate T cell development and functions remains unclear. We report that T cell-specific deletion of Keap1 results in defective development and reduced numbers of invariant natural killer T (NKT) cells in the thymus and the peripheral organs in a cell-intrinsic manner. The frequency of NKT2 and NKT17 cells increases while NKT1 decreases in these mice. Keap1-deficient NKT cells show increased rates of proliferation and apoptosis, as well as increased glucose uptake and mitochondrial function, but reduced ROS, CD122, and Bcl2 expression. In NKT cells deficient in Nrf2 and Keap1, all these phenotypic and metabolic defects are corrected. Thus, the Keap1-Nrf2 system contributes to NKT cell development and homeostasis by regulating cell metabolism.


Subject(s)
Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Natural Killer T-Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Thymus Gland/metabolism
9.
Proc Natl Acad Sci U S A ; 116(15): 7439-7448, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30910955

ABSTRACT

Cellular metabolism and signaling pathways are key regulators to determine conventional T cell fate and function, but little is understood about the role of cell metabolism for natural killer T (NKT) cell survival, proliferation, and function. We found that NKT cells operate distinct metabolic programming from CD4 T cells. NKT cells are less efficient in glucose uptake than CD4 T cells with or without activation. Gene-expression data revealed that, in NKT cells, glucose is preferentially metabolized by the pentose phosphate pathway and mitochondria, as opposed to being converted into lactate. In fact, glucose is essential for the effector functions of NKT cells and a high lactate environment is detrimental for NKT cell survival and proliferation. Increased glucose uptake and IFN-γ expression in NKT cells is inversely correlated with bacterial loads in response to bacterial infection, further supporting the significance of glucose metabolism for NKT cell function. We also found that promyelocytic leukemia zinc finger seemed to play a role in regulating NKT cells' glucose metabolism. Overall, our study reveals that NKT cells use distinct arms of glucose metabolism for their survival and function.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Mitochondria/metabolism , Natural Killer T-Cells/immunology , Oxidative Phosphorylation , Pentose Phosphate Pathway/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Survival/genetics , Cell Survival/immunology , Glucose/genetics , Glucose/immunology , Mice , Mice, Knockout , Mitochondria/genetics , Natural Killer T-Cells/cytology , Pentose Phosphate Pathway/genetics , Promyelocytic Leukemia Zinc Finger Protein/genetics , Promyelocytic Leukemia Zinc Finger Protein/immunology
10.
Eur J Immunol ; 48(7): 1255-1257, 2018 07.
Article in English | MEDLINE | ID: mdl-29572809

ABSTRACT

We show the presence of lymphoid tissue-resident PLZF+ CD45RA+ RO+ CD4 T cells in humans. They express HLA-DR, granzyme B, and perforin and are low on CCR7 like terminally differentiated effector memory (Temra) cells and are likely generated from effector T cells (Te) or from central (Tcm) or effector (Tem) memory T (Tcm) cells during immune responses. Tn, Naïve T cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Genotype , Lymphoid Tissue/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , T-Lymphocyte Subsets/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cells, Cultured , Granzymes/metabolism , HLA-DR Antigens/metabolism , Humans , Immunity, Cellular , Immunologic Memory , Perforin/metabolism
11.
Immune Netw ; 18(1): e14, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29503744

ABSTRACT

T lymphocytes rely on several metabolic processes to produce the high amounts of energy and metabolites needed to drive clonal expansion and the development of effector functions. However, many of these pathways result in the production of reactive oxygen species (ROS), which have canonically been thought of as cytotoxic agents due to their ability to damage DNA and other subcellular structures. Interestingly, ROS has recently emerged as a critical second messenger for T cell receptor signaling and T cell activation, but the sensitivity of different T cell subsets to ROS varies. Therefore, the tight regulation of ROS production by cellular antioxidant pathways is critical to maintaining proper signal transduction without compromising the integrity of the cell. This review intends to detail the common metabolic sources of intracellular ROS and the mechanisms by which ROS contributes to the development of T cell-mediated immunity. The regulation of ROS levels by the glutathione pathway and the Nrf2-Keap1-Cul3 trimeric complex will be discussed. Finally, T cell-mediated autoimmune diseases exacerbated by defects in ROS regulation will be further examined in order to identify potential therapeutic interventions for these disorders.

12.
J Immunol ; 199(10): 3478-3487, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29021374

ABSTRACT

Reactive oxygen species (ROS) are byproducts of aerobic metabolism and contribute to both physiological and pathological conditions as second messengers. ROS are essential for activation of T cells, but how ROS influence NKT cells is unknown. In the present study, we investigated the role of ROS in NKT cell function. We found that NKT cells, but not CD4 or CD8 T cells, have dramatically high ROS in the spleen and liver of mice but not in the thymus or adipose tissues. Accordingly, ROS-high NKT cells exhibited increased susceptibility and apoptotic cell death with oxidative stress. High ROS in the peripheral NKT cells were primarily produced by NADPH oxidases and not mitochondria. We observed that sorted ROS-high NKT cells were enriched in NKT1 and NKT17 cells, whereas NKT2 cells were dominant in ROS-low cells. Furthermore, treatment of NKT cells with antioxidants led to reduced frequencies of IFN-γ- and IL-17-expressing cells, indicating that ROS play a role in regulating the inflammatory function of NKT cells. The transcription factor promyelocytic leukemia zinc finger (PLZF) seemed to control the ROS levels. NKT cells from adipose tissues that do not express PLZF and those from PLZF haplodeficient mice have low ROS. Conversely, ROS were highly elevated in CD4 T cells from mice ectopically expressing PLZF. Thus, our findings demonstrate that PLZF controls ROS levels, which in turn governs the inflammatory function of NKT cells.


Subject(s)
Liver/immunology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Reactive Oxygen Species/metabolism , Spleen/immunology , Animals , Apoptosis , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidases/metabolism , Oxidative Stress , Promyelocytic Leukemia Zinc Finger Protein/genetics
13.
J Hepatol ; 67(1): 100-109, 2017 07.
Article in English | MEDLINE | ID: mdl-28267623

ABSTRACT

BACKGROUND & AIMS: The liver is an immunologically-privileged organ. Breakdown of liver immune privilege has been reported in chronic liver disease; however, the role of adaptive immunity in liver injury is poorly defined. Nuclear factor-κB-inducing kinase (NIK) is known to regulate immune tissue development, but its role in maintaining liver homeostasis remains unknown. This study aimed to assess the role of NIK, particularly thymic NIK, in regulating liver adaptive immunity. METHODS: NIK was deleted systemically or conditionally using the Cre/loxp system. Cluster of differentiation [CD]4+ or CD8+ T cells were depleted using anti-CD4 or anti-CD8 antibody. Donor bone marrows or thymi were transferred into recipient mice. Immune cells were assessed by immunohistochemistry and flow cytometry. RESULTS: Global, but not liver-specific or hematopoietic lineage cell-specific, deletion of NIK induced fatal liver injury, inflammation, and fibrosis. Likewise, adoptive transfer of NIK-null, but not wild-type, thymi into immune-deficient mice induced liver inflammation, injury, and fibrosis in recipients. Liver inflammation was characterized by a massive expansion of T cells, particularly the CD4+ T cell subpopulation. Depletion of CD4+, but not CD8+, T cells fully protected against liver injury, inflammation, and fibrosis in NIK-null mice. NIK deficiency also resulted in inflammation in the lung, kidney, and pancreas, but to a lesser degree relative to the liver. CONCLUSIONS: Thymic NIK suppresses development of autoreactive T cells against liver antigens, and NIK deficiency in the thymus results in CD4+ T cell-orchestrated autoimmune hepatitis and liver fibrosis. Thus, thymic NIK is essential for the maintenance of liver immune privilege and liver homeostasis. LAY SUMMARY: We found that global or thymus-specific ablation of the NIK gene results in fatal autoimmune liver disease in mice. NIK-deficient mice develop liver inflammation, injury, and fibrosis. Our findings indicate that thymic NIK is essential for the maintenance of liver integrity and homeostasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis, Autoimmune/etiology , Liver Cirrhosis, Experimental/etiology , Liver/immunology , Protein Serine-Threonine Kinases/physiology , Thymus Gland/physiology , Adaptive Immunity , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappaB-Inducing Kinase
14.
Mol Immunol ; 85: 47-56, 2017 05.
Article in English | MEDLINE | ID: mdl-28208073

ABSTRACT

The Wnt/ß-catenin signaling pathway plays important roles during various cellular functions including survival and proliferation of immune cells. The critical role of this pathway in conventional T cell development is established but little is known about its contributions to innate T cell development. In this study, we found that ß-catenin level, an indication of the strength of Wnt/ß-catenin signaling, is regulated during invariant NKT (iNKT) cell development. ß-catenin levels were greatly increased during iNKT cell selection from double positive thymocytes to Stage 0 of iNKT cell development and during subsequent development to Stage 1. Thereafter, ß-catenin levels decrease from Stage 2, which is essential for the terminal maturation of iNKT cells. Failure to dampen Wnt/ß-catenin signaling as in mice expressing a stabilized active form of ß-catenin (CATtg) resulted in increased Stage 2 and decreased Stage 3 iNKT cells. Inefficient transition from Stage 2 to 3 in CATtg iNKT cells seems to be contributed by poor expression of IL-15R (CD122) and transcription factor T-bet, both of which are necessary for terminal maturation of iNKT cells in the thymus. Consequently, IFN-γ+ iNKT cells were greatly reduced in CATtg mice. Together, our findings reveal that proper regulation of ß-catenin and in turn Wnt signaling plays an important role in the terminal maturation and function of iNKT cells.


Subject(s)
Cell Differentiation/immunology , Natural Killer T-Cells/immunology , Wnt Signaling Pathway/immunology , Animals , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction
15.
Leuk Lymphoma ; 57(2): 376-384, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26059058

ABSTRACT

Extracorporeal photopheresis (ECP) is a safe and effective immunoregulatory therapy for steroid-refractory chronic graft-versus-host disease (cGVHD) but its mechanism of action is poorly understood. In this study, we evaluated the effect of ECP in a sample of cGVHD patients. Our data showed that ECP-treated patients had lower CD4 T and B cells, and substantially higher NK cells than untreated patients. T regulatory (Treg) cells were similar between the two groups of patients. Interestingly, Treg cells were higher in ECP-treated patients and ECP-responders who had no history of aGVHD or sclerosis, than in those who had one of them or both. These findings suggest that at least one of the mechanisms of immunomodulation by ECP targets the Treg cell population and that an increase in Treg cells may be associated with response in patients with cGVHD. Together, the results of ECP are different depending on the patients' clinical condition.

16.
BMC Immunol ; 16: 62, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26482437

ABSTRACT

BACKGROUND: Invariant Natural Killer T (iNKT) cells have been implicated in lung inflammation in humans and also shown to be a key cell type in inducing allergic lung inflammation in mouse models. iNKT cells differentiate and acquire functional characteristics during development in the thymus. However, the correlation between development of iNKT cells in the thymus and role in lung inflammation remains unknown. In addition, transcriptional control of differentiation of iNKT cells into iNKT cell effector subsets in the thymus during development is also unclear. In this report we show that ß-catenin dependent mechanisms direct differentiation of iNKT2 and iNKT17 subsets but not iNKT1 cells. METHODS: To study the role for ß-catenin in lung inflammation we utilize mice with conditional deletion and enforced expression of ß-catenin in a well-established mouse model for IL-25-dependen lung inflammation. RESULTS: Specifically, we demonstrate that conditional deletion of ß-catenin permitted development of mature iNKT1 cells while impeding maturation of iNKT2 and 17 cells. A role for ß-catenin expression in promoting iNKT2 and iNKT17 subsets was confirmed when we noted that enforced transgenic expression of ß-catenin in iNKT cell precursors enhanced the frequency and number of iNKT2 and iNKT17 cells at the cost of iNKT1 cells. This effect of expression of ß-catenin in iNKT cell precursors was cell autonomous. Furthermore, iNKT2 cells acquired greater capability to produce type-2 cytokines when ß-catenin expression was enhanced. DISCUSSION: This report shows that ß-catenin deficiency resulted in a profound decrease in iNKT2 and iNKT17 subsets of iNKT cells whereas iNKT1 cells developed normally. By contrast, enforced expression of ß-catenin promoted the development of iNKT2 and iNKT17 cells. It was important to note that the majority of iNKT cells in the thymus of C57BL/6 mice were iNKT1 cells and enforced expression of ß-catenin altered the pattern to iNKT2 and iNKT17 cells suggesting that ß-catenin may be a major factor in the distinct pathways that critically direct differentiation of iNKT effector subsets. CONCLUSIONS: Thus, we demonstrate that ß-catenin expression in iNKT cell precursors promotes differentiation toward iNKT2 and iNKT17 effector subsets and supports enhanced capacity to produce type 2 and 17 cytokines which in turn augment lung inflammation in mice.


Subject(s)
Cell Differentiation , Interleukin-17/metabolism , Natural Killer T-Cells/immunology , Pneumonia/immunology , Pneumonia/pathology , beta Catenin/metabolism , Animals , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/complications
17.
J Immunol ; 194(1): 223-30, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25404366

ABSTRACT

The mammalian target of rapamycin (mTOR) senses and incorporates different environmental cues via the two signaling complexes mTOR complex 1 (mTORC1) and mTORC2. As a result, mTOR controls cell growth and survival, and also shapes different effector functions of the cells including immune cells such as T cells. We demonstrate in this article that invariant NKT (iNKT) cell development is controlled by mTORC2 in a cell-intrinsic manner. In mice deficient in mTORC2 signaling because of the conditional deletion of the Rictor gene, iNKT cell numbers were reduced in the thymus and periphery. This is caused by decreased proliferation of stage 1 iNKT cells and poor development through subsequent stages. Functionally, iNKT cells devoid of mTORC2 signaling showed reduced number of IL-4-expressing cells, which correlated with a decrease in the transcription factor GATA-3-expressing cells. However, promyelocytic leukemia zinc-finger (PLZF), a critical transcription factor for iNKT cell development, is expressed at a similar level in mTORC2-deficient iNKT cells compared with that in the wild type iNKT cells. Furthermore, cellular localization of PLZF was not altered in the absence of mTOR2 signaling. Thus, our study reveals the PLZF-independent mechanisms of the development and function of iNKT cells regulated by mTORC2.


Subject(s)
Carrier Proteins/immunology , GATA3 Transcription Factor/biosynthesis , Kruppel-Like Transcription Factors/biosynthesis , Multiprotein Complexes/immunology , Natural Killer T-Cells/cytology , TOR Serine-Threonine Kinases/immunology , Animals , Carrier Proteins/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-4/biosynthesis , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/genetics
18.
Immune Netw ; 14(1): 30-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24605078

ABSTRACT

Collaboration of TLR and non-TLR pathways in innate immune cells, which acts in concert for the induction of inflammatory cytokines, can mount a specific adaptive immune response tailored to a pathogen. Here, we show that murine DC produced increased IL-23 and IL-6 when they were treated with LPS together with curdlan that activates TLR4 and dectin-1, respectively. We also found that the induction of the inflammatory cytokine production by LPS and curdlan requires activation of IKK. However, the same treatment did not induce DC to produce a sufficient amount of TGF-ß. As a result, the conditioned media from DC treated with LPS and curdlan was not able to direct CD4(+) T cells to Th17 cells. Addition of TGF-ß but not IL-6 or IL-1ß was able to promote IL-17 production from CD4(+) T cells. Our results showed that although signaling mediated by LPS together with curdlan is a potent stimulator of DC to secrete many pro-inflammatory cytokines, TGF-ß production is a limiting factor for promoting Th17 immunity.

19.
J Immunol ; 191(2): 737-44, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23776174

ABSTRACT

MHC class II-expressing thymocytes can efficiently mediate positive selection of CD4 T cells in mice. Thymocyte-selected CD4 (T-CD4) T cells have an innate-like phenotype similar to invariant NKT cells. To investigate the development and function of T-CD4 T cells in-depth, we cloned TCR genes from T-CD4 T cells and generated transgenic mice. Remarkably, positive selection of T-CD4 TCR transgenic (T3) thymocytes occurred more efficiently when MHC class II was expressed by thymocytes than by thymic epithelial cells. Similar to polyclonal T-CD4 T cells and also invariant NKT cells, T3 CD4 T cell development is controlled by signaling lymphocyte activation molecule/signaling lymphocyte activation molecule-associated protein signaling, and the cells expressed both IL-4 and promyelocytic leukemia zinc finger (PLZF). Surprisingly, the selected T3 CD4 T cells were heterogeneous in that only half expressed IL-4 and only half expressed PLZF. IL-4- and PLZF-expressing cells were first found at the double-positive cell stage. Thus, the expression of IL-4 and PLZF seems to be determined by an unidentified event that occurs postselection and is not solely dependent on TCR specificity or the selection process, per se. Taken together, our data show for the first time, to our knowledge, that the TCR specificity regulates but does not determine the development of innate CD4 T cells by thymocytes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-4/metabolism , Kruppel-Like Transcription Factors/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/genetics , Animals , Antigens, CD/metabolism , Bone Marrow Cells , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chimera/genetics , Histocompatibility Antigens Class II , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/genetics , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Thymocytes/metabolism , Trans-Activators/genetics
20.
Proc Natl Acad Sci U S A ; 110(6): 2270-5, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23341605

ABSTRACT

Myeloid differentiation primary response protein 88 (MyD88) is classically known as an adaptor, linking TLR and IL-1R to downstream signaling pathways in the innate immune system. In addition to its role in innate immune cells, MyD88 has been shown to play an important role in T cells. How MyD88 regulates helper T-cell differentiation remains largely unknown, however. Here we demonstrate that MyD88 is an important regulator of IL-17-producing CD4(+) T helper cells (Th17) cell proliferation. MyD88-deficient CD4(+) T cells showed a defect in Th17 cell differentiation, but not in Th1 cell or Th2 cell differentiation. The impaired IL-17 production from MyD88-deficient CD4(+) T cells is not a result of defective RAR-related orphan receptor γt (RORγt) expression. Instead, MyD88 is essential for sustaining the mammalian target of rapamycin (mTOR) activation necessary to promote Th17 cell proliferation by linking IL-1 and IL-23 signaling. MyD88-deficient CD4(+) T cells showed impaired mTOR activation and, consequently, reduced Th17 cell proliferation. Importantly, the absence of MyD88 in T cells ameliorated disease in the experimental autoimmune encephalomyelitis model. Taken together, our results demonstrate that MyD88 has a dual function in Th17 cells by delivering IL-1 signaling during the early differentiation stage and integrating IL-23 signaling to the mTOR complex to expand committed Th17 cells.


Subject(s)
Interleukin-1/metabolism , Interleukin-23/metabolism , Myeloid Differentiation Factor 88/immunology , TOR Serine-Threonine Kinases/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immunity, Innate , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...