Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 23(2): 303-317, 2022 02.
Article in English | MEDLINE | ID: mdl-34949833

ABSTRACT

Antigen-specific memory CD4+ T cells can persist and confer rapid and efficient protection from microbial reinfection. However, the mechanisms underlying the long-term maintenance of the memory CD4+ T cell pool remain largely unknown. Here, using a mouse model of acute infection with lymphocytic choriomeningitis virus (LCMV), we found that the serine/threonine kinase complex mammalian target of rapamycin complex 2 (mTORC2) is critical for the long-term persistence of virus-specific memory CD4+ T cells. The perturbation of mTORC2 signaling at memory phase led to an enormous loss of virus-specific memory CD4+ T cells by a unique form of regulated cell death (RCD), ferroptosis. Mechanistically, mTORC2 inactivation resulted in the impaired phosphorylation of downstream AKT and GSK3ß kinases, which induced aberrant mitochondrial reactive oxygen species (ROS) accumulation and ensuing ferroptosis-causative lipid peroxidation in virus-specific memory CD4+ T cells; furthermore, the disruption of this signaling cascade also inhibited glutathione peroxidase 4 (GPX4), a major scavenger of lipid peroxidation. Thus, the mTORC2-AKT-GSK3ß axis functions as a key signaling hub to promote the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ferroptosis/immunology , Immunologic Memory/immunology , Longevity/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Animals , Glycogen Synthase Kinase 3 beta/immunology , Lipid Peroxidation/immunology , Lymphocyte Activation/immunology , Lymphocyte Count/methods , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/immunology
2.
Nat Commun ; 12(1): 2029, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795689

ABSTRACT

Mucosal-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 ß 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-1ß induces more MAIT17 than MAIT1 cells, while IL-23 alone promotes MAIT17 cell proliferation and survival, but synergizes with IL-1ß 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-1ßR/IL-23R to exert a crucial role for MAIT17 differentiation, while the IL-2/IL-15R-mTORC1-T-bet axis ensures MAIT1 differentiation.


Subject(s)
Cytokines/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-15/immunology , Interleukin-15/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucosal-Associated Invariant T Cells/cytology , Mucosal-Associated Invariant T Cells/metabolism , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
3.
Front Immunol ; 12: 582858, 2021.
Article in English | MEDLINE | ID: mdl-33679734

ABSTRACT

The structural and functional destruction of the blood-testis barrier (BTB) following uropathogenic E. coli (UPEC) infection may be a critical component of the pathologic progress of orchitis. Recent findings indicate that the mammalian target of the rapamycin (mTOR)-signaling pathway is implicated in the regulation of BTB assembly and restructuring. To explore the mechanisms underlying BTB damage induced by UPEC infection, we analyzed BTB integrity and the involvement of the mTOR-signaling pathway using in vivo and in vitro UPEC-infection models. We initially confirmed that soluble virulent factors secreted from UPEC trigger a stress response in Sertoli cells and disturb adjacent cell junctions via down-regulation of junctional proteins, including occludin, zonula occludens-1 (ZO-1), F-actin, connexin-43 (CX-43), ß-catenin, and N-cadherin. The BTB was ultimately disrupted in UPEC-infected rat testes, and blood samples from UPEC-induced orchitis in these animals were positive for anti-sperm antibodies. Furthermore, we herein also demonstrated that mTOR complex 1 (mTORC1) over-activation and mTORC2 suppression contributed to the disturbance in the balance between BTB "opening" and "closing." More importantly, rapamycin (a specific mTORC1 inhibitor) significantly restored the expression of cell-junction proteins and exerted a protective effect on the BTB during UPEC infection. We further confirmed that short-term treatment with rapamycin did not aggravate spermatogenic degeneration in infected rats. Collectively, this study showed an association between abnormal activation of the mTOR-signaling pathway and BTB impairment during UPEC-induced orchitis, which may provide new insights into a potential treatment strategy for testicular infection.


Subject(s)
Blood-Testis Barrier/immunology , Escherichia coli Infections/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Animals , Blood-Testis Barrier/metabolism , Cells, Cultured , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Orchitis/immunology , Orchitis/metabolism , Orchitis/microbiology , Rats, Sprague-Dawley , Sertoli Cells/immunology , Sertoli Cells/metabolism , Sertoli Cells/microbiology , Spermatogenesis/immunology , Testis/immunology , Testis/metabolism , Tight Junction Proteins/immunology , Tight Junction Proteins/metabolism , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/physiology
4.
Front Immunol ; 12: 751409, 2021.
Article in English | MEDLINE | ID: mdl-35069527

ABSTRACT

Dendritic cells (DCs) are recognized as a key orchestrator of immune response and homeostasis, deregulation of which may lead to autoimmunity such as experimental autoimmune encephalomyelitis (EAE). Herein we show that the phosphatase PP2Cδ played a pivotal role in regulating DC activation and function, as PP2Cδ ablation caused aberrant maturation, activation, and Th1/Th17-priming of DCs, and hence induced onset of exacerbated EAE. Mechanistically, PP2Cδ restrained the expression of the essential subunit of mTORC2, Rictor, primarily through de-phosphorylating and proteasomal degradation of the methyltransferase NSD2 via CRL4DCAF2 E3 ligase. Loss of PP2Cδ in DCs accordingly sustained activation of the Rictor/mTORC2 pathway and boosted glycolytic and mitochondrial metabolism. Consequently, ATP-citrate lyse (ACLY) was increasingly activated and catalyzed acetyl-CoA for expression of the genes compatible with hyperactivated DCs under PP2Cδ deletion. Collectively, our findings demonstrate that PP2Cδ has an essential role in controlling DCs activation and function, which is critical for prevention of autoimmunity.


Subject(s)
ATP Citrate (pro-S)-Lyase/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Histone-Lysine N-Methyltransferase/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Protein Phosphatase 2C/immunology , Signal Transduction/immunology , ATP Citrate (pro-S)-Lyase/genetics , Animals , Cell Differentiation/genetics , Female , Histone-Lysine N-Methyltransferase/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Knockout , Protein Phosphatase 2C/genetics , Signal Transduction/genetics
5.
J Immunol ; 205(9): 2456-2467, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32948684

ABSTRACT

Salmonella enterica serovar Typhimurium (S Typhimurium) is a Gram-negative bacterium that induces cell death of macrophages as a key virulence strategy. We have previously demonstrated that the induction of macrophage death is dependent on the host's type I IFN (IFN-I) response. IFN-I signaling has been shown to induce tripartite motif (TRIM) 21, an E3 ubiquitin ligase with critical functions in autoimmune disease and antiviral immunity. However, the importance and regulation of TRIM21 during bacterial infection remains poorly understood. In this study, we investigated the role of TRIM21 upon S Typhimurium infection of murine bone marrow-derived macrophages. Although Trim21 expression was induced in an IFN-I-dependent manner, we found that TRIM21 levels were mainly regulated posttranscriptionally. Following TLR4 activation, TRIM21 was transiently degraded via the lysosomal pathway by chaperone-mediated autophagy (CMA). However, S Typhimurium-induced mTORC2 signaling led to phosphorylation of Akt at S473, which subsequently impaired TRIM21 degradation by attenuating CMA. Elevated TRIM21 levels promoted macrophage death associated with reduced transcription of NF erythroid 2-related factor 2 (NRF2)-dependent antioxidative genes. Collectively, our results identify IFN-I-inducible TRIM21 as a negative regulator of innate immune responses to S Typhimurium and a previously unrecognized substrate of CMA. To our knowledge, this is the first study reporting that a member of the TRIM family is degraded by the lysosomal pathway.


Subject(s)
Chaperone-Mediated Autophagy/immunology , Ribonucleoproteins/immunology , Ribonucleoproteins/metabolism , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella typhimurium/immunology , Animals , Immunity, Innate/immunology , Lysosomes/immunology , Lysosomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 2/immunology , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/immunology
6.
Sci Rep ; 10(1): 10798, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612145

ABSTRACT

Celiac disease (CD) is an enteropathy triggered by the ingestion of gluten proteins in genetically predisposed individuals and characterized by excessive activation of effector immune cells and enhanced production of inflammatory cytokines. However, factors/mechanisms that amplify the ongoing mucosal inflammation in CD are not fully understood. In this study, we assessed whether mammalian target of Rapamycin (mTOR), a pathway that combines intra- and extra-cellular signals and acts as a central regulator for the metabolism, growth, and function of immune and non-immune cells, sustains CD-associated immune response. Our findings indicate that expression of phosphorylated (p)/active form of mTOR is increased in protein lysates of duodenal biopsy samples taken from patients with active CD (ACD) as compared to normal controls. In ACD, activation of mTOR occurs mainly in the epithelial compartment and associates with enhanced expression of p-4EBP, a downstream target of mTOR complex (mTORC)1, while expression of p-Rictor, a component of mTORC2, is not increased. Stimulation of mucosal explants of inactive CD patients with pepsin-trypsin-digested (PT)-gliadin or IFN-γ/IL-21, two cytokines produced in CD by gluten-specific T cells, increases p-4EBP expression. Consistently, blockade of such cytokines in cultures of ACD mucosal explants reduces p-4EBP. Finally, we show that inhibition of mTORC1 with rapamycin in ACD mucosal explants reduces p-4EBP and production of IL-15, a master cytokine produced by epithelial cells in this disorder. Our data suggest that ACD inflammation is marked by activation of mTORC1 in the epithelial compartment.


Subject(s)
Celiac Disease/immunology , Duodenum/immunology , Intestinal Mucosa/immunology , TOR Serine-Threonine Kinases/immunology , Biopsy , Celiac Disease/pathology , Duodenum/pathology , Female , Gliadin/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Interleukins/immunology , Intestinal Mucosa/pathology , Male , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Phosphorylation/immunology , T-Lymphocytes/immunology
7.
Front Immunol ; 11: 617404, 2020.
Article in English | MEDLINE | ID: mdl-33633735

ABSTRACT

The role of PI3K-mTOR pathway in regulating NK cell development has been widely reported. However, it remains unclear whether NK cell development depends on the protein kinase B (PKB), which links PI3K and mTOR, perhaps due to the potential redundancy of PKB. PKB has two phosphorylation sites, threonine 308 (T308) and serine 473 (S473), which can be phosphorylated by phosphoinositide-dependent protein kinase-1 (PDK1) and mTORC2, respectively. In this study, we established a mouse model in which PKB was inactivated through the deletion of PDK1 and Rictor, a key component of mTORC2, respectively. We found that the single deletion of PDK1 or Rictor could lead to a significant defect in NK cell development, while combined deletion of PDK1 and Rictor severely hindered NK cell development at the early stage. Notably, ectopic expression of myristoylated PKB significantly rescued this defect. In terms of mechanism, in PDK1/Rictor-deficient NK cells, E4BP4, a transcription factor for NK cell development, was less expressed, and the exogenous supply of E4BP4 could alleviate the developmental defect of NK cell in these mice. Besides, overexpression of Bcl-2 also helped the survival of PDK1/Rictor-deficient NK cells, suggesting an anti-apoptotic role of PKB in NK cells. In summary, complete phosphorylation of PKB at T308 and S473 by PDK1 and mTORC2 is necessary for optimal NK cell development, and PKB regulates NK cell development by promoting E4BP4 expression and preventing cell apoptosis.


Subject(s)
Killer Cells, Natural/immunology , Mechanistic Target of Rapamycin Complex 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Animals , Cell Differentiation/immunology , Enzyme Activation/immunology , Killer Cells, Natural/metabolism , Mechanistic Target of Rapamycin Complex 2/immunology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/immunology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/immunology , Signal Transduction/immunology
8.
Biochem Biophys Res Commun ; 521(1): 72-76, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31629474

ABSTRACT

Mast cells express high-affinity IgE receptor (FcεRI) on their surface, cross-linking of which leads to the immediate release of proinflammatory mediators such as histamine but also late-phase cytokine secretion, which are central to the pathogenesis of allergic diseases. Despite the growing evidences that mammalian target of rapamycin (mTOR) plays important roles in the immune system, it is still unclear how mTOR signaling regulates mast cell function. In this study, we investigated the effects of 3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-one (3BDO) as an mTOR agonist on FcεRI-mediated allergic responses of mast cells. Our data showed that administration of 3BDO decreased ß-hexosaminidase, interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α) release in murine bone marrow-derived mast cells (BMMCs) after FcεRI cross-linking, which was associated with an increase in mTOR complex 1 (mTORC1) signaling but a decrease in activation of Erk1/2, Jnk, and mTORC2-Akt. In addition, we found that a specific Akt agonist, SC79, is able to fully restore the decrease of ß-hexosaminidase release in 3BDO-treated BMMCs but has no effect on IL-6 release in these cells, suggesting that 3BDO negatively regulates FcεRI-mediated degranulation and cytokine release through differential mechanisms in mast cells. The present data demonstrate that proper activation of mTORC1 is crucial for mast cell effector function, suggesting the applicability of the mTORC1 activator as a useful therapeutic agent in mast cell-related diseases.


Subject(s)
4-Butyrolactone/analogs & derivatives , Cell Degranulation/drug effects , Mast Cells/drug effects , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, IgE/antagonists & inhibitors , 4-Butyrolactone/pharmacology , Animals , Mast Cells/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/immunology , Receptors, IgE/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
9.
Biochim Biophys Acta Mol Cell Res ; 1866(8): 1322-1337, 2019 08.
Article in English | MEDLINE | ID: mdl-30928610

ABSTRACT

The development of drug-resistance by neoplastic cells is recognized as a major cause of targeted therapy failure and disease progression. The mechanistic (previously mammalian) target of rapamycin (mTOR) is a highly conserved Ser/Thr kinase that acts as the catalytic subunit of two structurally and functionally distinct large multiprotein complexes, referred to as mTOR complex 1 (mTORC1) and mTORC2. Both mTORC1 and mTORC2 play key roles in a variety of healthy cell types/tissues by regulating physiological anabolic and catabolic processes in response to external cues. However, a body of evidence identified aberrant activation of mTOR signaling as a common event in many human tumors. Therefore, mTOR is an attractive target for therapeutic targeting in cancer and this fact has driven the development of numerous mTOR inhibitors, several of which have progressed to clinical trials. Nevertheless, mTOR inhibitors have met with a very limited success as anticancer therapeutics. Among other reasons, this failure was initially ascribed to the activation of several compensatory signaling pathways that dampen the efficacy of mTOR inhibitors. The discovery of these regulatory feedback mechanisms greatly contributed to a better understanding of cancer cell resistance to mTOR targeting agents. However, over the last few years, other mechanisms of resistance have emerged, including epigenetic alterations, compensatory metabolism rewiring and the occurrence of mTOR mutations. In this article, we provide the reader with an updated overview of the mechanisms that could explain resistance of cancer cells to the various classes of mTOR inhibitors.


Subject(s)
Neoplasm Proteins/immunology , Neoplasms/immunology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/immunology , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
10.
Am J Transplant ; 19(6): 1614-1621, 2019 06.
Article in English | MEDLINE | ID: mdl-30801921

ABSTRACT

A central role of the mechanistic target of rapamycin (mTOR) in regulation of fundamental cell processes is well recognized. mTOR functions in two distinct complexes: rapamycin-sensitive mTOR complex (C) 1 and rapamycin-insensitive mTORC2. While the role of mTORC1 in shaping immune responses, including transplant rejection, and the influence of its antagonism in promoting allograft tolerance have been studied extensively using rapamycin, lack of selective small molecule inhibitors has limited understanding of mTORC2 biology. Within the past few years, however, intracellular localization of mTORC2, its contribution to mitochondrial fitness, cell metabolism, cytoskeletal modeling and cell migration, and its role in differentiation and function of immune cells have been described. Studies in mTORC2 knockdown/knockout mouse models and a new class of dual mTORC1/2 inhibitors, have shed light on the immune regulatory functions of mTORC2. These include regulation of antigen-presenting cell, NK cell, T cell subset, and B cell differentiation and function. mTORC2 has been implicated in regulation of ischemia/reperfusion injury and graft rejection. Potential therapeutic benefits of antagonizing mTORC2 to inhibit chronic rejection have also been described, while selective in vivo targeting strategies using nanotechnology have been developed. We briefly review and discuss these developments and their implications.


Subject(s)
Mechanistic Target of Rapamycin Complex 2/immunology , Animals , B-Lymphocytes/immunology , Humans , Immunity, Innate , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mitochondria/metabolism , Models, Immunological , Reperfusion Injury/immunology , T-Lymphocyte Subsets/immunology , Transplantation Immunology
11.
Sci Signal ; 12(570)2019 02 26.
Article in English | MEDLINE | ID: mdl-30808817

ABSTRACT

Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that express an invariant T cell receptor (TCR), which recognizes glycolipid antigens presented on CD1d molecules. These cells are phenotypically and functionally distinct from conventional T cells. When we characterized the metabolic activity of iNKT cells, consistent with their activated phenotype, we found that they had much less mitochondrial respiratory capacity but increased glycolytic activity in comparison to naïve conventional CD4+ T cells. After TCR engagement, iNKT cells further increased aerobic glycolysis, which was important for the expression of interferon-γ (IFN-γ). Glycolytic metabolism promoted the translocation of hexokinase-II to mitochondria and the activation of mammalian target of rapamycin complex 2 (mTORC2). Inhibiting glycolysis reduced the activity of Akt and PKCθ, which inhibited TCR recycling and accumulation within the immune synapse. Diminished TCR accumulation in the immune synapse reduced the activation of proximal and distal TCR signaling pathways and IFN-γ production in activated iNKT cells. Our studies demonstrate that glycolytic metabolism augments TCR signaling duration and IFN-γ production in iNKT cells by increasing TCR recycling.


Subject(s)
Glycolysis/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Hexokinase/immunology , Hexokinase/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mechanistic Target of Rapamycin Complex 2/immunology , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/immunology , Mitochondria/metabolism , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/metabolism
12.
J Allergy Clin Immunol ; 143(6): 2038-2051.e12, 2019 06.
Article in English | MEDLINE | ID: mdl-30654047

ABSTRACT

BACKGROUND: Exposure to early-life undernutrition is closely related to higher risks of adverse immunologic outcomes in adulthood. Although it has been suggested that asthma has its origins in early life, its underlying mechanisms remain largely unknown. OBJECTIVE: We characterized the effects of early-life undernutrition on T lymphocytes, which play a pivotal role in immune diseases, and we investigated whether this contributes to susceptibility to asthma in adulthood. METHODS: Pregnant mice were fed a protein restriction diet (PRD) to establish an early-life undernutrition model. Naive CD4+ T cells (CD4+CD62LhiCD44-) from offspring were used throughout the study. TH2 differentiation was examined by using fluorescence-activated cell sorting and ELISA under TH2-polarized conditions in vitro and through ovalbumin-induced experimental asthma in vivo. T-cell metabolism was measured with a Seahorse XF96 Analyzer. DNA methylation levels were measured by using bisulfite sequencing. RESULTS: PRD CD4+ T cells displayed increased activation and proliferation and were prone to differentiate into TH2 cells both in vitro and in vivo, leading to susceptibility to experimental asthma. Mechanistically, early-life undernutrition upregulated mechanistic target of rapamycin 1-dependent glycolysis and induced conserved noncoding DNA sequence 1 DNA hypomethylation in the TH2 cytokine locus of CD4+ T cells. Glycolysis blockades undermined increased TH2 skewing and alleviated experimental asthma in PRD mice. CONCLUSION: Early-life undernutrition induced mechanistic target of rapamycin 1-dependent glycolysis upregulation and TH2 cytokine locus hypomethylation in CD4+ T cells, resulting in increased T-cell activation, proliferation, and TH2 skewing and further susceptibility to experimental asthma.


Subject(s)
Asthma/genetics , Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , Malnutrition/genetics , Malnutrition/immunology , Allergens/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Epigenesis, Genetic , Female , Glycolysis , Lung/immunology , Lung/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Mice, Inbred C57BL , Ovalbumin/immunology , Pregnancy
13.
Cell Immunol ; 335: 30-40, 2019 01.
Article in English | MEDLINE | ID: mdl-30420269

ABSTRACT

POTE is known as cancer antigen, expressed in many cancers, along with very few normal tissues like prostate, ovary, testes and embryo. Till date, POTEE identified as majorly expressed POTE paralog. Functionally, POTEF regulates TLR signaling which play important role in innate immunity provided clue about expression of POTE in immune cells. We have chosen three Thp1monocytes, Jurkat T1 and MΦ cells as a model. Here, first time we report expression of POTEE in immune cells specifically only in MΦ but not in monocytes or T-cells. In addition, expression level remains unaltered in MΦ subtypes M1 and M2 and MΦ subjected to various stresses, except MΦs treated with Hyp-CM where MΦs acquires properties of TAMs. In TAMs, POTEE was involved differential protein-protein interaction with mTOR, RICTOR, and Rad51 indicating its biological role in cell invasion through mTORC2 activation. siRNA mediated knockdown of POTEE suggests its importance in cell survival of MΦs as well as TAMs.


Subject(s)
Antigens, Neoplasm/biosynthesis , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Antigens, Neoplasm/immunology , Cell Movement/immunology , Cells, Cultured , Humans , Immunity, Innate , Jurkat Cells , Macrophages/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Monocytes/immunology , Monocytes/metabolism , Neoplasms/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , THP-1 Cells , Transcriptome
14.
Nat Immunol ; 19(10): 1071-1082, 2018 10.
Article in English | MEDLINE | ID: mdl-30201994

ABSTRACT

TLR3 is a sensor of double-stranded RNA that is indispensable for defense against infection with herpes simplex virus type 1 (HSV-1) in the brain. We found here that TLR3 was required for innate immune responses to HSV-1 in neurons and astrocytes. During infection with HSV-1, TLR3 recruited the metabolic checkpoint kinase complex mTORC2, which led to the induction of chemokines and trafficking of TLR3 to the cell periphery. Such trafficking enabled the activation of molecules (including mTORC1) required for the induction of type I interferons. Intracranial infection of mice with HSV-1 was exacerbated by impairment of TLR3 responses with an inhibitor of mTOR and was significantly 'rescued' by potentiation of TLR3 responses with an agonistic antibody to TLR3. These results suggest that the TLR3-mTORC2 axis might be a therapeutic target through which to combat herpes simplex encephalitis.


Subject(s)
Encephalitis, Herpes Simplex/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Toll-Like Receptor 3/immunology , Animals , Herpesvirus 1, Human , Immunity, Innate/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells
15.
Nat Commun ; 8: 14208, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128208

ABSTRACT

Alternatively activated macrophages (M2) have an important function in innate immune responses to parasitic helminths, and emerging evidence also indicates these cells are regulators of systemic metabolism. Here we show a critical role for mTORC2 signalling in the generation of M2 macrophages. Abrogation of mTORC2 signalling in macrophages by selective conditional deletion of the adaptor molecule Rictor inhibits the generation of M2 macrophages while leaving the generation of classically activated macrophages (M1) intact. Selective deletion of Rictor in macrophages prevents M2 differentiation and clearance of a parasitic helminth infection in mice, and also abrogates the ability of mice to regulate brown fat and maintain core body temperature. Our findings define a role for mTORC2 in macrophages in integrating signals from the immune microenvironment to promote innate type 2 immunity, and also to integrate systemic metabolic and thermogenic responses.


Subject(s)
Macrophages/physiology , Mechanistic Target of Rapamycin Complex 2/immunology , Strongylida Infections/immunology , Thermogenesis/physiology , Animals , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Female , Gene Knockout Techniques , Helminthiasis, Animal/immunology , Humans , Immunity, Innate/physiology , Male , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Nippostrongylus/immunology , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/immunology , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Signal Transduction/physiology , Strongylida Infections/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...