Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.514
Filter
1.
J Immunol ; 212(11): 1714-1721, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38598411

ABSTRACT

Ag-specific effector CD4+ T cells play a crucial role in defending against exogenous pathogens. However, the mechanisms governing the differentiation and function of IFN-γ-producing effector CD4+ Th1 cells in immune responses remain largely unknown. In this study, we elucidated the pivotal role of zinc finger protein 335 (Zfp335) in regulating effector Th1 cell differentiation and survival during acute bacterial infection. Mice with Zfp335 knockout in OT-II cells exhibited impaired Ag-specific CD4+ T cell expansion accompanied by a significant reduction in resistance to Listeria infection. Furthermore, Zfp335 deficiency restricted the effector CD4+ Th1 cell population and compromised their survival upon Listeria challenge. The expression of T-bet and IFN-γ was accordingly decreased in Zfp335-deficient Th1 cells. Mechanistically, Zfp335 directly bound to the promoter region of the Lmna gene and regulated its expression. Overexpression of Lmna was able to rescue the survival and function of Zfp335-deficient effector Th1 cells. Therefore, our study provides novel insights into the mechanisms governing effector Th1 cell differentiation and survival during acute infection.


Subject(s)
Cell Differentiation , DNA-Binding Proteins , Lamin Type A , Mice, Knockout , Th1 Cells , Transcription Factors , Animals , Mice , Cell Differentiation/immunology , Cell Differentiation/genetics , Th1 Cells/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lamin Type A/genetics , Mice, Inbred C57BL , Cell Survival/genetics , Cell Survival/immunology , Listeriosis/immunology , Gene Expression Regulation/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism
2.
Eur J Immunol ; 54(5): e2350873, 2024 May.
Article in English | MEDLINE | ID: mdl-38501878

ABSTRACT

Resident memory T (TRM) cells have been recently established as an important subset of memory T cells that provide early and essential protection against reinfection in the absence of circulating memory T cells. Recent findings showing that TRM expand in vivo after repeated antigenic stimulation indicate that these memory T cells are not terminally differentiated. This suggests an opportunity for in vitro TRM expansion to apply in an immunotherapy setting. However, it has also been shown that TRM may not maintain their identity and form circulating memory T cells after in vivo restimulation. Therefore, we set out to determine how TRM respond to antigenic activation in culture. Using Listeria monocytogenes and LCMV infection models, we found that TRM from the intraepithelial compartment of the small intestine expand in vitro after antigenic stimulation and subsequent resting in homeostatic cytokines. A large fraction of the expanded TRM retained their phenotype, including the expression of key TRM markers CD69 and CD103 (ITGAE). The optimal culture of TRM required low O2 pressure to maintain the expression of these and other TRM-associated molecules. Expanded TRM retained their effector capacity to produce cytokines after restimulation, but did not acquire a highly glycolytic profile indicative of effector T cells. The proteomic analysis confirmed TRM profile retention, including expression of TRM-related transcription factors, tissue retention factors, adhesion molecules, and enzymes involved in fatty acid metabolism. Collectively, our data indicate that limiting oxygen conditions supports in vitro expansion of TRM cells that maintain their TRM phenotype, at least in part, suggesting an opportunity for therapeutic strategies that require in vitro expansion of TRM.


Subject(s)
Immunologic Memory , Listeria monocytogenes , Memory T Cells , Animals , Memory T Cells/immunology , Immunologic Memory/immunology , Mice , Listeria monocytogenes/immunology , Antigens, CD/metabolism , Antigens, CD/immunology , Integrin alpha Chains/metabolism , Mice, Inbred C57BL , Listeriosis/immunology , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Cytokines/metabolism , Cytokines/immunology , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Intestinal Mucosa/immunology , CD8-Positive T-Lymphocytes/immunology , Intestine, Small/immunology , Cells, Cultured
3.
Foodborne Pathog Dis ; 21(5): 288-297, 2024 May.
Article in English | MEDLINE | ID: mdl-38237167

ABSTRACT

Gut microbiota (GM) has been proven to resist pathogenic infection through nutritional competition, colonization resistance and promotion of the host immune response. However, in clinical practice, GM is mainly used in intestinal diseases, such as Clostridium difficile infection, and there are few reports on its application in the treatment of pathogenic bacterial infections. In this study, GM from healthy mice was transplanted into mice infected with Listeria monocytogenes using fecal microbiota transplantation (FMT) and the effects were observed. We found that GM from healthy mice could reduce the mortality of infected mice and decrease the counts of L. monocytogenes in their liver and spleen. In addition, FMT inhibited the expression of inflammatory factors in the liver and spleen of infected mice. In vitro cell experiments revealed that GM can reduce the count of L. monocytogenes invading Caco-2 cells and inhibit the L. monocytogenes-caused apoptosis. These results indicate that GM can be used to protect mice infected with L. monocytogenes by eliminating the amount of L. monocytogenes in the host and inhibiting the overexpression of inflammatory factors. Hence, this method can potentially replace antibiotics in the treatment of L. monocytogenes infection.


Subject(s)
Apoptosis , Cytokines , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Listeria monocytogenes , Listeriosis , Animals , Listeriosis/microbiology , Listeriosis/immunology , Mice , Cytokines/metabolism , Humans , Caco-2 Cells , Liver/microbiology , Spleen/microbiology , Female
4.
Clin Immunol ; 257: 109819, 2023 12.
Article in English | MEDLINE | ID: mdl-37918467

ABSTRACT

Innate immune response is critical for the control of Listeria monocytogenes infection. Here, we identified developmentally regulated GTP-binding protein 2 (DRG2) in macrophages as a major regulator of the innate immune response against L. monocytogenes infection. Both whole-body DRG2 knockout (KO) mice and macrophage-specific DRG2 KO mice had low levels of IL-6 during early infection and increased susceptibility to L. monocytogenes infection. Following an initial impaired inflammatory response of macrophages upon i.p. L. monocytogenes infection, DRG2-/- mice showed delayed recruitment of neutrophils and monocytes into the peritoneal cavity, which led to elevated bacterial burden, inflammatory cytokine production at a late infection time point, and liver micro-abscesses. DRG2 deficiency decreased the transcriptional activity of NF-κB and impaired the inflammatory response of both bone marrow-derived and peritoneal macrophages upon L. monocytogenes stimulation. Our findings reveal that DRG2 in macrophages is critical for the initial inflammatory response and protection against L. monocytogenes infection.


Subject(s)
GTP-Binding Proteins , Listeria monocytogenes , Listeriosis , Macrophages , Animals , Mice , Immunity, Innate , Listeriosis/immunology , Macrophages/immunology , Mice, Knockout , Monocytes , GTP-Binding Proteins/metabolism
5.
Nature ; 606(7915): 769-775, 2022 06.
Article in English | MEDLINE | ID: mdl-35676476

ABSTRACT

Adaptive immune components are thought to exert non-overlapping roles in antimicrobial host defence, with antibodies targeting pathogens in the extracellular environment and T cells eliminating infection inside cells1,2. Reliance on antibodies for vertically transferred immunity from mothers to babies may explain neonatal susceptibility to intracellular infections3,4. Here we show that pregnancy-induced post-translational antibody modification enables protection against the prototypical intracellular pathogen Listeria monocytogenes. Infection susceptibility was reversed in neonatal mice born to preconceptually primed mothers possessing L. monocytogenes-specific IgG or after passive transfer of antibodies from primed pregnant, but not virgin, mice. Although maternal B cells were essential for producing IgGs that mediate vertically transferred protection, they were dispensable for antibody acquisition of protective function, which instead required sialic acid acetyl esterase5 to deacetylate terminal sialic acid residues on IgG variable-region N-linked glycans. Deacetylated L. monocytogenes-specific IgG protected neonates through the sialic acid receptor CD226,7, which suppressed IL-10 production by B cells leading to antibody-mediated protection. Consideration of the maternal-fetal dyad as a joined immunological unit reveals protective roles for antibodies against intracellular infection and fine-tuned adaptations to enhance host defence during pregnancy and early life.


Subject(s)
Immunity, Maternally-Acquired , Immunoglobulin G , Intracellular Space , Listeria monocytogenes , Mothers , Pregnancy , Acetylesterase , Animals , Animals, Newborn , B-Lymphocytes , Female , Immunity, Maternally-Acquired/immunology , Immunoglobulin G/immunology , Interleukin-10/biosynthesis , Intracellular Space/immunology , Intracellular Space/microbiology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/prevention & control , Mice , N-Acetylneuraminic Acid/metabolism , Pregnancy/immunology , Sialic Acid Binding Ig-like Lectin 2 , T-Lymphocytes
6.
Int Immunopharmacol ; 108: 108742, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35378447

ABSTRACT

BACKGROUND: Interleukin-33 (IL-33) is a member of the interleukin-1 family, which is reported to be important across a range of diseases. However, the mechanisms underlying IL-33/ST2 axis in infectious diseases have not yet been fully addressed. METHODS: We established both lipopolysaccharide (LPS)-induced injuryin T cells and Listeria monocytogenes (Lm) infection model to determine the effect of IL-33 on infectious immunity. RESULTS: The T cell proliferation was inhibited by LPS while IL-33 could reverse the outcome. Further, apoptosis was significantly promoted after serum stimulation (ST)2 knockdown, suggesting IL-33, acting through its receptor ST2, may attenuate the inhibitory effect of LPS on T cells through the apoptotic signaling pathway. In this study, we also identified an IL-33-mediated mechanism of T cell differentiation in pregnant mice infected with Lm. Here, we observed the elevated expression of IL-33 in pregnant mice infected with Lm. Furthermore, we revealed that blocking IL-33 markedly decreased the abortion rate and placental bacterial load, but weakened placental inflammatory repair, by inhibiting Th2 cell-mediated immune responses and relatively intensifying Th1-dominent immunoreaction. CONCLUSIONS: These findings reveal a previously unidentified mechanism underlying IL-33/ST2 axis. IL-33 signaling and targeting T cell-mediated immunity may present a new therapeutic strategy for the treatment of infectious diseases.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Listeria , Listeriosis , T-Lymphocytes , Animals , Female , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics , Lipopolysaccharides , Listeriosis/immunology , Lymphocyte Activation , Mice , Placenta , Pregnancy , T-Lymphocytes/immunology
7.
Proc Natl Acad Sci U S A ; 119(10): e2113329119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35239442

ABSTRACT

SignificanceThe CD4+ Treg response following acute Listeria infection is heterogeneous and deploys two distinct modes of suppression coinciding with initial pathogen exposure and resolution of infection. This bimodal suppression of CD8+ T cells during priming and contraction is mediated by separate Treg lineages. These findings make a significant contribution to our understanding of the functional plasticity inherent within Tregs, which allows these cells to serve as a sensitive and dynamic cellular rheostat for the immune system to prevent autoimmune pathology in the face of inflammation attendant to acute infection, enable expansion of the pathogen-specific response needed to control the infection, and reestablish immune homeostasis after the threat has been contained.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/immunology , Acute Disease , Animals , Mice
8.
Science ; 375(6583): 859-863, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35201883

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are innate immune effectors that contribute to host defense. Whether ILC3 functions are stably modified after pathogen encounter is unknown. Here, we assess the impact of a time-restricted enterobacterial challenge to long-term ILC3 activation in mice. We found that intestinal ILC3s persist for months in an activated state after exposure to Citrobacter rodentium. Upon rechallenge, these "trained" ILC3s proliferate, display enhanced interleukin-22 (IL-22) responses, and have a superior capacity to control infection compared with naïve ILC3s. Metabolic changes occur in C. rodentium-exposed ILC3s, but only trained ILC3s have an enhanced proliferative capacity that contributes to increased IL-22 production. Accordingly, a limited encounter with a pathogen can promote durable phenotypic and functional changes in intestinal ILC3s that contribute to long-term mucosal defense.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Lymphocyte Activation , Lymphocytes/immunology , Adaptive Immunity , Animals , Cell Proliferation , Female , Immunity, Innate , Immunologic Memory , Interleukins/metabolism , Intestines/immunology , Listeria monocytogenes , Listeriosis/immunology , Lymphocytes/metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Oxygen Consumption , RNA-Seq , Reinfection/immunology , Interleukin-22
9.
J Immunol ; 208(5): 1155-1169, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35110421

ABSTRACT

CD8+ T cells are critical for the immune response to pathogens and tumors, and CD8+ T cell memory protects against repeat infections. In this study, we identify the activating transcription factor 7 interacting protein (ATF7ip) as a critical regulator of CD8+ T cell immune responses. Mice with a T cell-specific deletion of ATF7ip have a CD8+ T cell-intrinsic enhancement of Il7r expression and Il2 expression leading to enhanced effector and memory responses. Chromatin immunoprecipitation sequencing studies identified ATF7ip as a repressor of Il7r and Il2 gene expression through the deposition of the repressive histone mark H3K9me3 at the Il7r gene and Il2-Il21 intergenic region. Interestingly, ATF7ip targeted transposable elements for H3K9me3 deposition at both the IL7r locus and the Il2-Il21 intergenic region, indicating that ATF7ip silencing of transposable elements is important for regulating CD8+ T cell function. These results demonstrate a new epigenetic pathway by which IL-7R and IL-2 production are constrained in CD8+ T cells, and this may open up new avenues for modulating their production.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-2/biosynthesis , Receptors, Interleukin-7/biosynthesis , Repressor Proteins/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Chromatin Immunoprecipitation , DNA Transposable Elements/genetics , Gene Deletion , Gene Silencing , Histones/genetics , Humans , Interleukin-2/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Repressor Proteins/genetics
10.
Immunity ; 55(2): 254-271.e7, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139352

ABSTRACT

Allergic immunity is orchestrated by group 2 innate lymphoid cells (ILC2s) and type 2 helper T (Th2) cells prominently arrayed at epithelial- and microbial-rich barriers. However, ILC2s and Th2 cells are also present in fibroblast-rich niches within the adventitial layer of larger vessels and similar boundary structures in sterile deep tissues, and it remains unclear whether they undergo dynamic repositioning during immune perturbations. Here, we used thick-section quantitative imaging to show that allergic inflammation drives invasion of lung and liver non-adventitial parenchyma by ILC2s and Th2 cells. However, during concurrent type 1 and type 2 mixed inflammation, IFNγ from broadly distributed type 1 lymphocytes directly blocked both ILC2 parenchymal trafficking and subsequent cell survival. ILC2 and Th2 cell confinement to adventitia limited mortality by the type 1 pathogen Listeria monocytogenes. Our results suggest that the topography of tissue lymphocyte subsets is tightly regulated to promote appropriately timed and balanced immunity.


Subject(s)
Inflammation/immunology , Interferon-gamma/immunology , Lymphocyte Subsets/immunology , Th2 Cells/immunology , Animals , Cell Death/immunology , Cell Movement/immunology , Hypersensitivity/immunology , Immunity, Innate , Interleukin-33/immunology , Interleukin-5/metabolism , Listeria monocytogenes , Listeriosis/immunology , Listeriosis/mortality , Liver/immunology , Lung/immunology , Lymphocyte Subsets/metabolism , Lysophospholipids/immunology , Mice , Parenchymal Tissue/immunology , Sphingosine/analogs & derivatives , Sphingosine/immunology , Th1 Cells/immunology , Th2 Cells/metabolism
11.
Int J Mol Sci ; 23(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35216061

ABSTRACT

Listeria monocytogenes (Lm) bacterial ghosts (LMGs) were produced by the minimum inhibitory concentration (MIC) of HCl, H2SO4, and NaOH. Acid and alkali effects on the LMGs were compared by in vitro and in vivo analyses. Scanning electron microscope showed that all chemicals form lysis pores on the Lm cell envelopes. Real-time qPCR revealed a complete absence of genomic DNA in HCl- and H2SO4-induced LMGs but not in NaOH-induced LMGs. HCl-, H2SO4- and NaOH-induced LMGs showed weaker or missing protein bands on SDS-PAGE gel when compared to wild-type Lm. Murine macrophages exposed to the HCl-induced LMGs showed higher cell viability than those exposed to NaOH-induced LMGs or wild-type Lm. The maximum level of cytokine expression (TNF-α, iNOS, IFN-γ, and IL-10 mRNA) was observed in the macrophages exposed to NaOH-induced LMGs, while that of IL-1ß mRNA was observed in the macrophages exposed to HCl-induced LMGs. To investigate LMGs as a vaccine candidate, mice were divided into PBS buffer-injected, HCl- and NaOH-induced LMGs immunized groups. Mice vaccinated with HCl- and NOH-induced LMGs, respectively, significantly increased in specific IgG antibodies, bactericidal activities of serum, and CD4+ and CD8+ T-cell population. Antigenic Lm proteins reacted with antisera against HCl- and NOH-induced LMGs, respectively. Bacterial loads in HCl- and NaOH-induced LMGs immunized mice were significantly lower than PBS-injected mice after virulent Lm challenges. It suggested that vaccination with LMGs induces both humoral and cell-mediated immune responses and protects against virulent challenges.


Subject(s)
Hydrochloric Acid/immunology , Immunity, Cellular/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Sodium Hydroxide/immunology , Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cytokines/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Rats
12.
Mol Cell ; 82(3): 527-541.e7, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35016033

ABSTRACT

Citrulline can be converted into argininosuccinate by argininosuccinate synthetase (ASS1) in the urea cycle and the citrulline-nitric oxide cycle. However, the regulation and biological function of citrulline metabolism remain obscure in the immune system. Unexpectedly, we found that macrophage citrulline declines rapidly after interferon gamma (IFN-γ) and/or lipopolysaccharide (LPS) stimulation, which is required for efficient proinflammatory signaling activation. Mechanistically, IFN-γ and/or LPS stimulation promotes signal transducers and activators of transcription 1 (STAT1)-mediated ASS1 transcription and Janus kinase2 (JAK2)-mediated phosphorylation of ASS1 at tyrosine 87, thereby leading to citrulline depletion. Reciprocally, increased citrulline directly binds to JAK2 and inhibits JAK2-STAT1 signaling. Blockage of ASS1-mediated citrulline depletion suppresses the host defense against bacterial infection in vivo. We therefore define a central role for ASS1 in controlling inflammatory macrophage activation and antibacterial defense through depletion of cellular citrulline and, further, identify citrulline as an innate immune-signaling metabolite that engages a metabolic checkpoint for proinflammatory responses.


Subject(s)
Argininosuccinate Synthase/metabolism , Citrulline/metabolism , Immunity, Innate , Inflammation/enzymology , Listeriosis/enzymology , Macrophage Activation , Macrophages/enzymology , Animals , Argininosuccinate Synthase/genetics , Disease Models, Animal , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Listeria monocytogenes/immunology , Listeriosis/genetics , Listeriosis/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RAW 264.7 Cells , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction
13.
Immunol Invest ; 51(4): 1005-1022, 2022 May.
Article in English | MEDLINE | ID: mdl-33830841

ABSTRACT

Macrophages are crucial effectors of innate immunity against the pathogenic bacterium Listeria monocytogenes. The pro-inflammatory cytokine tumour necrosis factor-α (TNF) has been shown to be crucial for resistance to L. monocytogenes and mice deficient in TNF signalling succumb quickly after infection. However, the mechanisms underlying TNF-mediated defence against L. monocytogenes infection have not been completely elucidated. Here, we demonstrate that TNF concurrently functions to support a pro-inflammatory M1 phenotype while actively blocking macrophage polarization to the M2 phenotype. Compared to WT mice, peritoneal macrophages in TNF-deficient mice inoculated with L. monocytogenes respond with M2 polarization by upregulating Arg1. Consistently, TNF blockade in vitro resulted in M2 polarization in peritoneal macrophages during L. monocytogenes infection. Additionally, TNF promotes the transition from M2 to M1 polarization in peritoneal macrophages. Further investigation of peritoneal macrophage polarization suggested the NF-κB pathway is involved in the TNF-dependent M2 to M1 shift. Conversely, treatment of peritoneal macrophage with a PPARγ agonist blunted the expression of M1 genes induced by TNF and reduced NF-κB signalling pathway activation. Competing signalling mechanisms therefore play an essential role in the ability of peritoneal macrophage to resolve L. monocytogenes infections with TNF playing an essential role in driving M1 polarization.Abbreviations: LPM: large peritoneal macrophage; SPM: small peritoneal macrophage; LLO: listeriolysin O; iNOS: inducible nitric oxide synthase; DCs: dendritic cells.


Subject(s)
Listeriosis , Macrophage Activation , Macrophages, Peritoneal , Tumor Necrosis Factor-alpha , Animals , Listeriosis/immunology , Macrophages , Macrophages, Peritoneal/metabolism , Mice , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/genetics
14.
J Immunol ; 208(1): 155-168, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34872976

ABSTRACT

CD8+ memory T (TM) cells play a critical role in immune defense against infection. Two common γ-chain family cytokines, IL-2 and IL-7, although triggering the same mTORC1-S6K pathway, distinctly induce effector T (TE) cells and TM cells, respectively, but the underlying mechanism(s) remains elusive. In this study, we generated IL-7R-/and AMPKα1-knockout (KO)/OTI mice. By using genetic and pharmaceutical tools, we demonstrate that IL-7 deficiency represses expression of FOXO1, TCF1, p-AMPKα1 (T172), and p-ULK1 (S555) and abolishes T cell memory differentiation in IL-7R KO T cells after Listeria monocytogenesis rLmOVA infection. IL-2- and IL-7-stimulated strong and weak S6K (IL-2/S6Kstrong and IL-7/S6Kweak) signals control short-lived IL-7R-CD62L-KLRG1+ TE and long-term IL-7R+CD62L+KLRG1- TM cell formations, respectively. To assess underlying molecular pathway(s), we performed flow cytometry, Western blotting, confocal microscopy, and Seahorse assay analyses by using the IL-7/S6Kweak-stimulated TM (IL-7/TM) and the control IL-2/S6Kstrong-stimulated TE (IL-2/TE) cells. We determine that the IL-7/S6Kweak signal activates transcriptional FOXO1, TCF1, and Id3 and metabolic p-AMPKα1, p-ULK1, and ATG7 molecules in IL-7/TM cells. IL-7/TM cells upregulate IL-7R and CD62L, promote mitochondria biogenesis and fatty acid oxidation metabolism, and show long-term cell survival and functional recall responses. Interestingly, AMPKα1 deficiency abolishes the AMPKα1 but maintains the FOXO1 pathway and induces a metabolic switch from fatty acid oxidation to glycolysis in AMPKα1 KO IL-7/TM cells, leading to loss of cell survival and recall responses. Taken together, our data demonstrate that IL-7-stimulated weak strength of mTORC1-S6K signaling controls T cell memory via activation of transcriptional FOXO1-TCF1-Id3 and metabolic AMPKα1-ULK1-ATG7 pathways. This (to our knowledge) novel finding provides a new mechanism for a distinct IL-2/IL-7 stimulation model in T cell memory and greatly impacts vaccine development.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , CD8-Positive T-Lymphocytes/immunology , Forkhead Box Protein O1/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Inhibitor of Differentiation Proteins/metabolism , Interleukin-7/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Memory T Cells/immunology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Cytotoxicity, Immunologic , Fatty Acids/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Regulation , Glycolysis , Hepatocyte Nuclear Factor 1-alpha/genetics , Immunologic Memory , Inhibitor of Differentiation Proteins/genetics , Interleukin-7/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/genetics , Signal Transduction , Vaccine Development
15.
J Immunol ; 208(1): 97-109, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34872978

ABSTRACT

Dendritic cell (DC) activation is characterized by sustained commitment to glycolysis that is a requirement for survival in DC subsets that express inducible NO synthase (Nos2) due to NO-mediated inhibition of mitochondrial respiration. This phenomenon primarily has been studied in DCs from the classic laboratory inbred mouse strain C57BL/6J (B6) mice, where DCs experience a loss of mitochondrial function due to NO accumulation. To assess the conservation of NO-driven metabolic regulation in DCs, we compared B6 mice to the wild-derived genetically divergent PWD/PhJ (PWD) strain. We show preserved mitochondrial respiration and enhanced postactivation survival due to attenuated NO production in LPS-stimulated PWD DCs phenocopying human monocyte-derived DCs. To genetically map this phenotype, we used a congenic mouse strain (B6.PWD-Chr11.2) that carries a PWD-derived portion of chromosome 11, including Nos2, on a B6 background. B6.PWD-Chr11.2 DCs show preserved mitochondrial function and produce lower NO levels than B6 DCs. We demonstrate that activated B6.PWD-Chr11.2 DCs maintain mitochondrial respiration and TCA cycle carbon flux, compared with B6 DCs. However, reduced NO production by the PWD Nos2 allele results in impaired cellular control of Listeria monocytogenes replication. These studies establish a natural genetic model for restrained endogenous NO production to investigate the contribution of NO in regulating the interplay between DC metabolism and immune function. These findings suggest that reported differences between human and murine DCs may be an artifact of the limited genetic diversity of the mouse models used, underscoring the need for mouse genetic diversity in immunology research.


Subject(s)
Dendritic Cells/immunology , Listeria monocytogenes/physiology , Listeriosis/immunology , Mitochondria/metabolism , Nitric Oxide/metabolism , Alleles , Animals , Animals, Wild , Cell Survival , Cells, Cultured , Disease Models, Animal , Disease Resistance , Genetic Background , Humans , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism
16.
Front Immunol ; 12: 793918, 2021.
Article in English | MEDLINE | ID: mdl-34956227

ABSTRACT

Interleukin-15, produced by hematopoietic and parenchymal cells, maintains immune cell homeostasis and facilitates activation of lymphoid and myeloid cell subsets. IL-15 interacts with the ligand-binding receptor chain IL-15Rα during biosynthesis, and the IL-15:IL-15Rα complex is trans-presented to responder cells that express the IL-2/15Rßγc complex to initiate signaling. IL-15-deficient and IL-15Rα-deficient mice display similar alterations in immune cell subsets. Thus, the trimeric IL-15Rαßγc complex is considered the functional IL-15 receptor. However, studies on the pathogenic role of IL-15 in inflammatory and autoimmune diseases indicate that IL-15 can signal independently of IL-15Rα via the IL-15Rßγc dimer. Here, we compared the ability of mice lacking IL-15 (no signaling) or IL-15Rα (partial/distinct signaling) to control Listeria monocytogenes infection. We show that IL-15-deficient mice succumb to infection whereas IL-15Rα-deficient mice clear the pathogen as efficiently as wildtype mice. IL-15-deficient macrophages did not show any defect in bacterial uptake or iNOS expression in vitro. In vivo, IL-15 deficiency impaired the accumulation of inflammatory monocytes in infected spleens without affecting chemokine and pro-inflammatory cytokine production. The inability of IL-15-deficient mice to clear L. monocytogenes results from impaired early IFNγ production, which was not affected in IL-15Rα-deficient mice. Administration of IFNγ partially enabled IL-15-deficient mice to control the infection. Bone marrow chimeras revealed that IL-15 needed for early bacterial control can originate from both hematopoietic and non-hematopoietic cells. Overall, our findings indicate that IL-15-dependent IL-15Rα-independent signaling via the IL-15Rßγc dimeric complex is necessary and sufficient for the induction of IFNγ from sources other than NK/NKT cells to control bacterial pathogens.


Subject(s)
Interferon-gamma/metabolism , Interleukin-15/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/immunology , Receptors, Interleukin-15/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Interferon-gamma/genetics , Interleukin-15/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phagocytosis , Receptors, Interleukin-15/genetics , Signal Transduction , Transplantation Chimera
17.
Cell Rep ; 37(5): 109956, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731605

ABSTRACT

Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Liver/immunology , Malaria/immunology , Plasmodium berghei/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/parasitology , Disease Models, Animal , Female , Host-Parasite Interactions , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/blood , Listeriosis/immunology , Listeriosis/microbiology , Liver/metabolism , Liver/microbiology , Liver/parasitology , Lymphocyte Function-Associated Antigen-1/metabolism , Malaria/blood , Malaria/parasitology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Parasite Load , Phagocytes/immunology , Phagocytes/metabolism , Phagocytes/microbiology , Phagocytes/parasitology , Plasmodium berghei/pathogenicity , Time Factors
18.
Cell Rep ; 37(6): 109969, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758312

ABSTRACT

MicroRNAs (miRNAs) have emerged as critical regulators of cell fate in the CD8+ T cell response to infection. Although there are several examples of miRNAs acting on effector CD8+ T cells after infection, it is unclear whether differential expression of one or more miRNAs in the naive state is consequential in altering their long-term trajectory. To answer this question, we examine the role of miR-29 in neonatal and adult CD8+ T cells, which express different amounts of miR-29 only prior to infection and adopt profoundly different fates after immune challenge. We find that manipulation of miR-29 expression in the naive state is sufficient for age-adjusting the phenotype and function of CD8+ T cells, including their regulatory landscapes and long-term differentiation trajectories after infection. Thus, miR-29 acts as a developmental switch by controlling the balance between a rapid effector response in neonates and the generation of long-lived memory in adults.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Listeriosis/immunology , Lymphocyte Activation/immunology , MicroRNAs/genetics , Adolescent , Adult , Age Factors , Animals , CD8-Positive T-Lymphocytes/microbiology , Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Humans , Listeria monocytogenes/immunology , Listeriosis/microbiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Young Adult
19.
Front Immunol ; 12: 723393, 2021.
Article in English | MEDLINE | ID: mdl-34603295

ABSTRACT

Trained immunity refers to the ability of the innate immune system exposed to a first challenge to provide an enhanced response to a secondary homologous or heterologous challenge. We reported that training induced with ß-glucan one week before infection confers protection against a broad-spectrum of lethal bacterial infections. Whether this protection persists over time is unknown. To tackle this question, we analyzed the immune status and the response to Listeria monocytogenes (L. monocytogenes) of mice trained 9 weeks before analysis. The induction of trained immunity increased bone marrow myelopoiesis and blood counts of Ly6Chigh inflammatory monocytes and polymorphonuclear neutrophils (PMNs). Ex vivo, whole blood, PMNs and monocytes from trained mice produced increased levels of cytokines in response to microbial products and limited the growth of L. monocytogenes. In vivo, following challenge with L. monocytogenes, peripheral blood leukocytes were massively depleted in control mice but largely preserved in trained mice. PMNs were reduced also in the spleen from control mice, and increased in the spleen of trained mice. In transwell experiments, PMNs from trained mice showed increased spontaneous migration and CXCL2/MIP2α-induced chemotaxis, suggesting that training promotes the migration of PMNs in peripheral organs targeted by L. monocytogenes. Trained PMNs and monocytes had higher glycolytic activity and mitochondrial respiration than control cells when exposed to L. monocytogenes. Bacterial burden and dissemination in blood, spleen and liver as well as systemic cytokines and inflammation (multiplex bead assay and bioluminescence imaging) were reduced in trained mice. In full agreement with these results, mice trained 9 weeks before infection were powerfully protected from lethal listeriosis. Altogether, these data suggest that training increases the generation and the antimicrobial activity of PMNs and monocytes, which may confer prolonged protection from lethal bacterial infection.


Subject(s)
Immunity, Innate , Listeria monocytogenes/immunology , Listeriosis/immunology , Monocytes/metabolism , Neutrophils/metabolism , Animals , Bone Marrow , Cytokines/metabolism , Female , Inflammation/immunology , Listeriosis/microbiology , Mice , Mice, Inbred C57BL
20.
Front Immunol ; 12: 724855, 2021.
Article in English | MEDLINE | ID: mdl-34659211

ABSTRACT

The adhesion and degranulation-promoting adaptor protein (ADAP) serves as a multifunctional scaffold and is involved in the formation of immune signaling complexes. To date, only limited data exist regarding the role of ADAP in pathogen-specific immunity during in vivo infection, and its contribution in phagocyte-mediated antibacterial immunity remains elusive. Here, we show that mice lacking ADAP (ADAPko) are highly susceptible to the infection with the intracellular pathogen Listeria monocytogenes (Lm) by showing enhanced immunopathology in infected tissues together with increased morbidity, mortality, and excessive infiltration of neutrophils and monocytes. Despite high phagocyte numbers in the spleen and liver, ADAPko mice only inefficiently controlled pathogen growth, hinting at a functional impairment of infection-primed phagocytes in the ADAP-deficient host. Flow cytometric analysis of hallmark pro-inflammatory mediators and unbiased whole genome transcriptional profiling of neutrophils and inflammatory monocytes uncovered broad molecular alterations in the inflammatory program in both phagocyte subsets following their activation in the ADAP-deficient host. Strikingly, ex vivo phagocytosis assay revealed impaired phagocytic capacity of neutrophils derived from Lm-infected ADAPko mice. Together, our data suggest that an alternative priming of phagocytes in ADAP-deficient mice during Lm infection induces marked alterations in the inflammatory profile of neutrophils and inflammatory monocytes that contribute to enhanced immunopathology while limiting their capacity to eliminate the pathogen and to prevent the fatal outcome of the infection.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Phagocytes/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Female , Immunity , Listeriosis/metabolism , Listeriosis/microbiology , Liver/metabolism , Male , Mice , Mice, Knockout , Phagocytes/metabolism , Phenotype , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...