Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.516
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Article in English | MEDLINE | ID: mdl-34663978

ABSTRACT

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Heredity , Immunity, Innate/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Myeloid Cells/immunology , Animals , Candida albicans/pathogenicity , Candidiasis/genetics , Candidiasis/metabolism , Candidiasis/microbiology , Cells, Cultured , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Male , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Spermatozoa/immunology , Spermatozoa/metabolism , Transcription, Genetic
2.
Nat Immunol ; 22(6): 699-710, 2021 06.
Article in English | MEDLINE | ID: mdl-34040226

ABSTRACT

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeriosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Proliferation/genetics , Colitis/microbiology , Colitis/pathology , Colon/cytology , Colon/embryology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Germ-Free Life , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/pathology , Listeriosis/microbiology , Listeriosis/pathology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
3.
Cell ; 175(6): 1651-1664.e14, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30392956

ABSTRACT

The activator and composition of the NLRP6 inflammasome remain poorly understood. We find that lipoteichoic acid (LTA), a molecule produced by Gram-positive bacteria, binds and activates NLRP6. In response to cytosolic LTA or infection with Listeria monocytogenes, NLRP6 recruited caspase-11 and caspase-1 via the adaptor ASC. NLRP6 activation by LTA induced processing of caspase-11, which promoted caspase-1 activation and interleukin-1ß (IL-1ß)/IL-18 maturation in macrophages. Nlrp6-/- and Casp11-/- mice were less susceptible to L. monocytogenes infection, which was associated with reduced pathogen loads and impaired IL-18 production. Administration of IL-18 to Nlrp6-/- or Casp11-/- mice restored the susceptibility of mutant mice to L. monocytogenes infection. These results reveal a previously unrecognized innate immunity pathway triggered by cytosolic LTA that is sensed by NLRP6 and exacerbates systemic Gram-positive pathogen infection via the production of IL-18.


Subject(s)
Immunity, Innate , Inflammasomes/immunology , Lipopolysaccharides/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Receptors, Cell Surface/immunology , Teichoic Acids/immunology , Animals , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Listeriosis/genetics , Listeriosis/pathology , Mice , Mice, Knockout , Receptors, Cell Surface/genetics
4.
Cell ; 171(5): 1057-1071.e11, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29033131

ABSTRACT

Type I interferon restrains interleukin-1ß (IL-1ß)-driven inflammation in macrophages by upregulating cholesterol-25-hydroxylase (Ch25h) and repressing SREBP transcription factors. However, the molecular links between lipid metabolism and IL-1ß production remain obscure. Here, we demonstrate that production of 25-hydroxycholesterol (25-HC) by macrophages is required to prevent inflammasome activation by the DNA sensor protein absent in melanoma 2 (AIM2). We find that in response to bacterial infection or lipopolysaccharide (LPS) stimulation, macrophages upregulate Ch25h to maintain repression of SREBP2 activation and cholesterol synthesis. Increasing macrophage cholesterol content is sufficient to trigger IL-1ß release in a crystal-independent but AIM2-dependent manner. Ch25h deficiency results in cholesterol-dependent reduced mitochondrial respiratory capacity and release of mitochondrial DNA into the cytosol. AIM2 deficiency rescues the increased inflammasome activity observed in Ch25h-/-. Therefore, activated macrophages utilize 25-HC in an anti-inflammatory circuit that maintains mitochondrial integrity and prevents spurious AIM2 inflammasome activation.


Subject(s)
Cholesterol/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Animals , Cholesterol/biosynthesis , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Humans , Hydroxycholesterols/metabolism , Inflammasomes/immunology , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/cytology , Macrophages/immunology , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Oxysterols/metabolism
5.
Nat Immunol ; 20(12): 1621-1630, 2019 12.
Article in English | MEDLINE | ID: mdl-31740800

ABSTRACT

Interferon-γ (IFN-γ) is essential for the innate immune response to intracellular bacteria. Noncoding RNAs and RNA-binding proteins (RBPs) need to be further considered in studies of regulation of the IFN-γ-activated signaling pathway in macrophages. In the present study, we found that the microRNA miR-1 promoted IFN-γ-mediated clearance of Listeria monocytogenes in macrophages by indirectly stabilizing the Stat1 messenger RNA through the degradation of the cytoplasmic long noncoding RNA Sros1. Inducible degradation or genetic loss of Sros1 led to enhanced IFN-γ-dependent activation of the innate immune response. Mechanistically, Sros1 blocked the binding of Stat1 mRNA to the RBP CAPRIN1, which stabilized the Stat1 mRNA and, consequently, promoted IFN-γ-STAT1-mediated innate immunity. These observations shed light on the complex RNA-RNA regulatory networks involved in cytokine-initiated innate responses in host-pathogen interactions.


Subject(s)
Cytoplasm/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/immunology , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , STAT1 Transcription Factor/metabolism , Animals , Cell Cycle Proteins/metabolism , Immunity, Innate , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Protein Binding , RAW 264.7 Cells , RNA Stability , RNA, Long Noncoding/metabolism , STAT1 Transcription Factor/genetics
6.
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
7.
Cell ; 165(7): 1672-1685, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27315481

ABSTRACT

Long intergenic noncoding RNAs (lincRNAs) are important regulators of gene expression. Although lincRNAs are expressed in immune cells, their functions in immunity are largely unexplored. Here, we identify an immunoregulatory lincRNA, lincRNA-EPS, that is precisely regulated in macrophages to control the expression of immune response genes (IRGs). Transcriptome analysis of macrophages from lincRNA-EPS-deficient mice, combined with gain-of-function and rescue experiments, revealed a specific role for this lincRNA in restraining IRG expression. Consistently, lincRNA-EPS-deficient mice manifest enhanced inflammation and lethality following endotoxin challenge in vivo. lincRNA-EPS localizes at regulatory regions of IRGs to control nucleosome positioning and repress transcription. Further, lincRNA-EPS mediates these effects by interacting with heterogeneous nuclear ribonucleoprotein L via a CANACA motif located in its 3' end. Together, these findings identify lincRNA-EPS as a repressor of inflammatory responses, highlighting the importance of lincRNAs in the immune system.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Macrophages/immunology , RNA, Long Noncoding/metabolism , Animals , Chromatids/metabolism , Gene Deletion , Humans , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/virology , Mice , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , Respirovirus Infections/immunology , Sendai virus/physiology , Toll-Like Receptors/metabolism , Transcriptome
8.
Nat Immunol ; 19(2): 141-150, 2018 02.
Article in English | MEDLINE | ID: mdl-29292386

ABSTRACT

Cyclic diadenylate monophosphate (c-di-AMP) is secreted by bacteria as a secondary messenger. How immune cells detect c-di-AMP and initiate anti-bacterial immunity remains unknown. We found that the endoplasmic reticulum (ER) membrane adaptor ERAdP acts as a direct sensor for c-di-AMP. ERAdP-deficient mice were highly susceptible to Listeria monocytogenes infection and exhibited reduced pro-inflammatory cytokines. Mechanistically, c-di-AMP bound to the C-terminal domain of ERAdP, which in turn led to dimerization of ERAdP, resulting in association with and activation of the kinase TAK1. TAK1 activation consequently initiated activation of the transcription factor NF-κB to induce the production of pro-inflammatory cytokines in innate immune cells. Moreover, double-knockout of ERAdP and TAK1 resulted in heightened susceptibility to L. monocytogenes infection. Thus, ERAdP-mediated production of pro-inflammatory cytokines is critical for controlling bacterial infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Dinucleoside Phosphates/immunology , Immunity, Innate/immunology , Listeriosis/immunology , Membrane Proteins/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Second Messenger Systems/immunology
9.
Immunity ; 54(4): 829-844.e5, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33705706

ABSTRACT

Memory T cells are thought to rely on oxidative phosphorylation and short-lived effector T cells on glycolysis. Here, we investigated how T cells arrive at these states during an immune response. To understand the metabolic state of rare, early-activated T cells, we adapted mass cytometry to quantify metabolic regulators at single-cell resolution in parallel with cell signaling, proliferation, and effector function. We interrogated CD8+ T cell activation in vitro and in response to Listeria monocytogenes infection in vivo. This approach revealed a distinct metabolic state in early-activated T cells characterized by maximal expression of glycolytic and oxidative metabolic proteins. Cells in this transient state were most abundant 5 days post-infection before rapidly decreasing metabolic protein expression. Analogous findings were observed in chimeric antigen receptor (CAR) T cells interrogated longitudinally in advanced lymphoma patients. Our study demonstrates the utility of single-cell metabolic analysis by mass cytometry to identify metabolic adaptations of immune cell populations in vivo and provides a resource for investigations of metabolic regulation of immune responses across a variety of applications.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , Animals , Cell Proliferation/physiology , Female , Glycolysis/immunology , Immunologic Memory/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Phosphorylation , Receptors, Chimeric Antigen/immunology , Single-Cell Analysis/methods
10.
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
11.
Nat Immunol ; 18(5): 573-582, 2017 05.
Article in English | MEDLINE | ID: mdl-28288100

ABSTRACT

Dynamic changes in the expression of transcription factors (TFs) can influence the specification of distinct CD8+ T cell fates, but the observation of equivalent expression of TFs among differentially fated precursor cells suggests additional underlying mechanisms. Here we profiled the genome-wide histone modifications, open chromatin and gene expression of naive, terminal-effector, memory-precursor and memory CD8+ T cell populations induced during the in vivo response to bacterial infection. Integration of these data suggested that the expression and binding of TFs contributed to the establishment of subset-specific enhancers during differentiation. We developed a new bioinformatics method using the PageRank algorithm to reveal key TFs that influence the generation of effector and memory populations. The TFs YY1 and Nr3c1, both constitutively expressed during CD8+ T cell differentiation, regulated the formation of terminal-effector cell fates and memory-precursor cell fates, respectively. Our data define the epigenetic landscape of differentiation intermediates and facilitate the identification of TFs with previously unappreciated roles in CD8+ T cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Epigenesis, Genetic , Listeriosis/immunology , Receptors, Glucocorticoid/metabolism , T-Lymphocyte Subsets/physiology , YY1 Transcription Factor/metabolism , Animals , CD8-Positive T-Lymphocytes/microbiology , Cell Differentiation/genetics , Computational Biology , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Histones/metabolism , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL , Receptors, Glucocorticoid/genetics , T-Lymphocyte Subsets/microbiology , YY1 Transcription Factor/genetics
12.
Nat Immunol ; 17(4): 379-86, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901151

ABSTRACT

The T cell antigen receptor (TCR) is unique in that its affinity for ligand is unknown before encounter and can vary by orders of magnitude. How the immune system regulates individual T cells that display very different reactivity to antigen remains unclear. Here we found that activated CD4(+) T cells, at the peak of clonal expansion, persistently downregulated their TCR expression in proportion to the strength of the initial antigen recognition. This programmed response increased the threshold for cytokine production and recall proliferation in a clone-specific manner and ultimately excluded clones with the highest antigen reactivity. Thus, programmed downregulation of TCR expression represents a negative feedback mechanism for constraining T cell effector function with a suitable time delay to thereby allow pathogen control while avoiding excess inflammatory damage.


Subject(s)
Down-Regulation , Listeriosis/immunology , Receptors, Antigen, T-Cell/genetics , Th1 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Immunoblotting , Listeria monocytogenes , Lymphocyte Activation , Mice , Mice, Transgenic , Mycobacterium tuberculosis , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes , Transcriptome
13.
Nat Immunol ; 17(10): 1167-75, 2016 10.
Article in English | MEDLINE | ID: mdl-27548433

ABSTRACT

CD8α(+) dendritic cells (DCs) are specialized at cross-presenting extracellular antigens on major histocompatibility complex (MHC) class I molecules to initiate cytotoxic T lymphocyte (CTL) responses; however, details of the mechanisms that regulate cross-presentation remain unknown. We found lower expression of the lectin family member Siglec-G in CD8α(+) DCs, and Siglec-G deficient (Siglecg(-/-)) mice generated more antigen-specific CTLs to inhibit intracellular bacterial infection and tumor growth. MHC class I-peptide complexes were more abundant on Siglecg(-/-) CD8α(+) DCs than on Siglecg(+/+) CD8α(+) DCs. Mechanistically, phagosome-expressed Siglec-G recruited the phosphatase SHP-1, which dephosphorylated the NADPH oxidase component p47(phox) and inhibited the activation of NOX2 on phagosomes. This resulted in excessive hydrolysis of exogenous antigens, which led to diminished formation of MHC class I-peptide complexes for cross-presentation. Therefore, Siglec-G inhibited DC cross-presentation by impairing such complex formation, and our results add insight into the regulation of cross-presentation in adaptive immunity.


Subject(s)
Cross-Priming , Dendritic Cells/immunology , Lectins/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Neoplasms, Experimental/immunology , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens/metabolism , CD8 Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Lectins/genetics , Lymphocyte Activation , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/metabolism , Peptide Fragments/metabolism , Phagocytosis/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Immunoglobulin-like Lectins , Signal Transduction , Tumor Burden/genetics
14.
Immunity ; 51(1): 64-76.e7, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31231033

ABSTRACT

Type 1 CD8α+ conventional dendritic cells (cDC1s) are required for CD8+ T cell priming but, paradoxically, promote splenic Listeria monocytogenes infection. Using mice with impaired cDC2 function, we ruled out a role for cDC2s in this process and instead discovered an interleukin-10 (IL-10)-dependent cellular crosstalk in the marginal zone (MZ) that promoted bacterial infection. Mice lacking the guanine nucleotide exchange factor DOCK8 or CD19 lost IL-10-producing MZ B cells and were resistant to Listeria. IL-10 increased intracellular Listeria in cDC1s indirectly by reducing inducible nitric oxide synthase expression early after infection and increasing intracellular Listeria in MZ metallophilic macrophages (MMMs). These MMMs trans-infected cDC1s, which, in turn, transported Listeria into the white pulp to prime CD8+ T cells. However, this also facilitated bacterial expansion. Therefore, IL-10-mediated crosstalk between B cells, macrophages, and cDC1s in the MZ promotes both Listeria infection and CD8+ T cell activation.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Interleukin-10/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/immunology , Spleen/immunology , Animals , Antigens, CD19/metabolism , CD8 Antigens/metabolism , Cell Line, Tumor , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/genetics , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Paracrine Communication , Spleen/microbiology
15.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746840

ABSTRACT

A "switch" from oxidative phosphorylation (OXPHOS) to aerobic glycolysis is a hallmark of T cell activation and is thought to be required to meet the metabolic demands of proliferation. However, why proliferating cells adopt this less efficient metabolism, especially in an oxygen-replete environment, remains incompletely understood. We show here that aerobic glycolysis is specifically required for effector function in T cells but that this pathway is not necessary for proliferation or survival. When activated T cells are provided with costimulation and growth factors but are blocked from engaging glycolysis, their ability to produce IFN-γ is markedly compromised. This defect is translational and is regulated by the binding of the glycolysis enzyme GAPDH to AU-rich elements within the 3' UTR of IFN-γ mRNA. GAPDH, by engaging/disengaging glycolysis and through fluctuations in its expression, controls effector cytokine production. Thus, aerobic glycolysis is a metabolically regulated signaling mechanism needed to control cellular function.


Subject(s)
Glycolysis , Lymphocyte Activation , Oxidative Phosphorylation , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , 3' Untranslated Regions , Animals , Cell Proliferation , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Interferon-gamma/genetics , Listeria monocytogenes , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Protein Biosynthesis , T-Lymphocytes/immunology
16.
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
17.
Nat Immunol ; 16(9): 918-26, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26237551

ABSTRACT

Mucosal immunity protects a host from intestinal inflammation and infection and is profoundly influenced by symbiotic bacteria. Here we report that in mice symbiotic bacteria directed selective cargo sorting in Paneth cells to promote symbiosis through Nod2, a cytosolic bacterial sensor, and the multifunctional protein kinase LRRK2, both encoded by inflammatory bowel disease (IBD)-associated genes. Commensals recruited Nod2 onto lysozyme-containing dense core vesicles (DCVs), which was required for DCV localization of LRRK2 and a small GTPase, Rab2a. Deficiency of Nod2, LRRK2 or Rab2a or depletion of commensals resulted in lysosomal degradation of lysozyme. Thus, commensal bacteria and host factors orchestrate the lysozyme-sorting process to protect the host from enteric infection, implicating Paneth cell dysfunction in IBD pathogenesis.


Subject(s)
Enterocolitis/immunology , Immunity, Mucosal/immunology , Inflammatory Bowel Diseases/immunology , Intestines/immunology , Listeriosis/immunology , Paneth Cells/immunology , Protein Serine-Threonine Kinases/immunology , Symbiosis/immunology , Animals , Enterocolitis/genetics , Immunity, Mucosal/genetics , Inflammatory Bowel Diseases/genetics , Intestines/microbiology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Listeriosis/genetics , Lysosomes , Mice , Mice, Knockout , Muramidase , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Protein Serine-Threonine Kinases/genetics , Secretory Vesicles/immunology , Symbiosis/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/immunology
18.
Nat Immunol ; 16(5): 525-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25848866

ABSTRACT

Innate immunological signals induced by pathogen- and/or damage-associated molecular patterns are essential for adaptive immune responses, but it is unclear if the brain has a role in this process. Here we found that while the abundance of tumor-necrosis factor (TNF) quickly increased in the brain of mice following bacterial infection, intra-brain delivery of TNF mimicked bacterial infection to rapidly increase the number of peripheral lymphocytes, especially in the spleen and fat. Studies of various mouse models revealed that hypothalamic responses to TNF were accountable for this increase in peripheral lymphocytes in response to bacterial infection. Finally, we found that hypothalamic induction of lipolysis mediated the brain's action in promoting this increase in the peripheral adaptive immune response. Thus, the brain-fat axis is important for rapid linkage of innate immunity to adaptive immunity.


Subject(s)
Adipose Tissue/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hypothalamus/immunology , Listeriosis/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/microbiology , Cell Count , Cells, Cultured , Fatty Acids/blood , Hypothalamus/microbiology , Immunity, Innate , Lipolysis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/cerebrospinal fluid
19.
Nat Immunol ; 16(4): 366-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25729924

ABSTRACT

Neutrophils express Toll-like receptors (TLRs) for the recognition of conserved bacterial elements to initiate antimicrobial responses. However, whether other cytosolic DNA sensors are expressed by neutrophils remains elusive. Here we found constitutive expression of the transcription factor Sox2 in the cytoplasm of mouse and human neutrophils. Neutrophil-specific Sox2 deficiency exacerbated bacterial infection. Sox2 directly recognized microbial DNA through its high-mobility-group (HMG) domain. Upon challenge with bacterial DNA, Sox2 dimerization was needed to activate a complex of the kinase TAK1 and its binding partner TAB2, which led to activation of the transcription factors NF-κB and AP-1 in neutrophils. Deficiency in TAK1 or TAB2 impaired Sox2-mediated antibacterial immunity. Overall, we reveal a previously unrecognized role for Sox2 as a cytosolic sequence-specific DNA sensor in neutrophils, which might provide potential therapeutic strategies for the treatment of infectious diseases.


Subject(s)
DNA, Bacterial/immunology , Immunity, Innate , Listeriosis/immunology , Neutrophils/immunology , SOXB1 Transcription Factors/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Cytoplasm/immunology , Cytoplasm/microbiology , Gene Expression Regulation , Humans , Listeria monocytogenes/immunology , Listeriosis/genetics , Listeriosis/microbiology , Listeriosis/mortality , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Mice , NF-kappa B/genetics , NF-kappa B/immunology , Neutrophils/microbiology , Protein Multimerization , SOXB1 Transcription Factors/genetics , Signal Transduction , Survival Analysis , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology
20.
Nat Immunol ; 15(3): 266-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487322

ABSTRACT

Interactions of T cell antigen receptors (TCRs) with complexes of self peptide and major histocompatibility complex (MHC) are crucial to T cell development, but their role in peripheral T cell responses remains unclear. Specific and nonspecific stimulation of LLO56 and LLO118 T cells, which transgenically express a TCR specific for the same Listeria monocytogenes epitope, elicited distinct interleukin 2 (IL-2) and phosphorylated kinase Erk responses, the strength of which was set in the thymus and maintained in the periphery in proportion to the avidity of the binding of the TCR to the self peptide-MHC complex. Deprivation of self peptide-MHC substantially compromised the population expansion of LLO56 T cells in response to L. monocytogenes in vivo. Despite their very different self-reactivity, LLO56 T cells and LLO118 T cells bound cognate peptide-MHC with an identical affinity, which challenges associations made between these parameters. Our findings highlight a crucial role for selecting ligands encountered during thymic 'education' in determining the intrinsic functionality of CD4+ T cells.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , Adoptive Transfer , Animals , Cell Separation , Flow Cytometry , Humans , Immunoblotting , Listeriosis/immunology , Mice , Mice, Knockout , Surface Plasmon Resonance , Thymus Gland/cytology , Thymus Gland/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL