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










Publication year range
1.
Nature ; 620(7973): 374-380, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532932

ABSTRACT

Low-grade inflammation is a hallmark of old age and a central driver of ageing-associated impairment and disease1. Multiple factors can contribute to ageing-associated inflammation2; however, the molecular pathways that transduce aberrant inflammatory signalling and their impact in natural ageing remain unclear. Here we show that the cGAS-STING signalling pathway, which mediates immune sensing of DNA3, is a critical driver of chronic inflammation and functional decline during ageing. Blockade of STING suppresses the inflammatory phenotypes of senescent human cells and tissues, attenuates ageing-related inflammation in multiple peripheral organs and the brain in mice, and leads to an improvement in tissue function. Focusing on the ageing brain, we reveal that activation of STING triggers reactive microglial transcriptional states, neurodegeneration and cognitive decline. Cytosolic DNA released from perturbed mitochondria elicits cGAS activity in old microglia, defining a mechanism by which cGAS-STING signalling is engaged in the ageing brain. Single-nucleus RNA-sequencing analysis of microglia and hippocampi of a cGAS gain-of-function mouse model demonstrates that engagement of cGAS in microglia is sufficient to direct ageing-associated transcriptional microglial states leading to bystander cell inflammation, neurotoxicity and impaired memory capacity. Our findings establish the cGAS-STING pathway as a driver of ageing-related inflammation in peripheral organs and the brain, and reveal blockade of cGAS-STING signalling as a potential strategy to halt neurodegenerative processes during old age.


Subject(s)
Aging , Brain , Cognitive Dysfunction , Inflammation , Membrane Proteins , Neurodegenerative Diseases , Nucleotidyltransferases , Animals , Humans , Mice , Aging/metabolism , Aging/pathology , Brain/metabolism , Brain/pathology , Bystander Effect , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , DNA/immunology , Inflammation/enzymology , Inflammation/metabolism , Membrane Proteins/metabolism , Memory Disorders/enzymology , Memory Disorders/metabolism , Microglia/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/metabolism , Nucleotidyltransferases/metabolism , Organ Specificity , Signal Transduction , Hippocampus/metabolism , Hippocampus/pathology
2.
Nature ; 603(7899): 145-151, 2022 03.
Article in English | MEDLINE | ID: mdl-35045565

ABSTRACT

COVID-19, which is caused by infection with SARS-CoV-2, is characterized by lung pathology and extrapulmonary complications1,2. Type I interferons (IFNs) have an essential role in the pathogenesis of COVID-19 (refs 3-5). Although rapid induction of type I IFNs limits virus propagation, a sustained increase in the levels of type I IFNs in the late phase of the infection is associated with aberrant inflammation and poor clinical outcome5-17. Here we show that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which controls immunity to cytosolic DNA, is a critical driver of aberrant type I IFN responses in COVID-19 (ref. 18). Profiling COVID-19 skin manifestations, we uncover a STING-dependent type I IFN signature that is primarily mediated by macrophages adjacent to areas of endothelial cell damage. Moreover, cGAS-STING activity was detected in lung samples from patients with COVID-19 with prominent tissue destruction, and was associated with type I IFN responses. A lung-on-chip model revealed that, in addition to macrophages, infection with SARS-CoV-2 activates cGAS-STING signalling in endothelial cells through mitochondrial DNA release, which leads to cell death and type I IFN production. In mice, pharmacological inhibition of STING reduces severe lung inflammation induced by SARS-CoV-2 and improves disease outcome. Collectively, our study establishes a mechanistic basis of pathological type I IFN responses in COVID-19 and reveals a principle for the development of host-directed therapeutics.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Interferon Type I/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , SARS-CoV-2/immunology , Animals , COVID-19/metabolism , COVID-19/virology , Cells, Cultured , DNA, Mitochondrial/metabolism , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Female , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Macrophages/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/virology , SARS-CoV-2/pathogenicity , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology
3.
Open Biol ; 11(3): 210030, 2021 03.
Article in English | MEDLINE | ID: mdl-33715389

ABSTRACT

Nucleic acid sensing through pattern recognition receptors is critical for immune recognition of microbial infections. Microbial DNA is frequently methylated at the N6 position of adenines (m6A), a modification that is rare in mammalian host DNA. We show here how that m6A methylation of 5'-GATC-3' motifs augments the immunogenicity of synthetic double-stranded (ds)DNA in murine macrophages and dendritic cells. Transfection with m6A-methylated DNA increased the expression of the activation markers CD69 and CD86, and of Ifnß, iNos and Cxcl10 mRNA. Similar to unmethylated cytosolic dsDNA, recognition of m6A DNA occurs independently of TLR and RIG-I signalling, but requires the two key mediators of cytosolic DNA sensing, STING and cGAS. Intriguingly, the response to m6A DNA is sequence-specific. m6A is immunostimulatory in some motifs, but immunosuppressive in others, a feature that is conserved between mouse and human macrophages. In conclusion, epigenetic alterations of DNA depend on the context of the sequence and are differentially perceived by innate cells, a feature that could potentially be used for the design of immune-modulating therapeutics.


Subject(s)
Adenine/analogs & derivatives , DNA Methylation , Immunity, Innate , Oligodeoxyribonucleotides/immunology , Adenine/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , B7-2 Antigen/metabolism , Cells, Cultured , Chemokine CXCL10/metabolism , Cytoplasm/metabolism , Dendritic Cells/immunology , Humans , Interferon-beta/metabolism , Lectins, C-Type/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oligodeoxyribonucleotides/chemistry , Toll-Like Receptors/metabolism
4.
PLoS Pathog ; 14(12): e1007491, 2018 12.
Article in English | MEDLINE | ID: mdl-30571761

ABSTRACT

The ESX-1, type VII, secretion system represents the major virulence determinant of Mycobacterium tuberculosis, one of the most successful intracellular pathogens. Here, by combining genetic and high-throughput approaches, we show that EspL, a protein of 115 amino acids, is essential for mediating ESX-1-dependent virulence and for stabilization of EspE, EspF and EspH protein levels. Indeed, an espL knock-out mutant was unable to replicate intracellularly, secrete ESX-1 substrates or stimulate innate cytokine production. Moreover, proteomic studies detected greatly reduced amounts of EspE, EspF and EspH in the espL mutant as compared to the wild type strain, suggesting a role for EspL as a chaperone. The latter conclusion was further supported by discovering that EspL interacts with EspD, which was previously demonstrated to stabilize the ESX-1 substrates and effector proteins, EspA and EspC. Loss of EspL also leads to downregulation in M. tuberculosis of WhiB6, a redox-sensitive transcriptional activator of ESX-1 genes. Overall, our data highlight the importance of a so-far overlooked, though conserved, component of the ESX-1 secretion system and begin to delineate the role played by EspE, EspF and EspH in virulence and host-pathogen interaction.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/pathogenicity , Virulence Factors/metabolism , Virulence/physiology , Humans , Mycobacterium tuberculosis/metabolism , THP-1 Cells , Tuberculosis/microbiology
5.
Nature ; 559(7713): 269-273, 2018 07.
Article in English | MEDLINE | ID: mdl-29973723

ABSTRACT

Aberrant activation of innate immune pathways is associated with a variety of diseases. Progress in understanding the molecular mechanisms of innate immune pathways has led to the promise of targeted therapeutic approaches, but the development of drugs that act specifically on molecules of interest remains challenging. Here we report the discovery and characterization of highly potent and selective small-molecule antagonists of the stimulator of interferon genes (STING) protein, which is a central signalling component of the intracellular DNA sensing pathway1,2. Mechanistically, the identified compounds covalently target the predicted transmembrane cysteine residue 91 and thereby block the activation-induced palmitoylation of STING. Using these inhibitors, we show that the palmitoylation of STING is essential for its assembly into multimeric complexes at the Golgi apparatus and, in turn, for the recruitment of downstream signalling factors. The identified compounds and their derivatives reduce STING-mediated inflammatory cytokine production in both human and mouse cells. Furthermore, we show that these small-molecule antagonists attenuate pathological features of autoinflammatory disease in mice. In summary, our work uncovers a mechanism by which STING can be inhibited pharmacologically and demonstrates the potential of therapies that target STING for the treatment of autoinflammatory disease.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Binding Sites , Cell Line , Cysteine/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Hereditary Autoinflammatory Diseases/drug therapy , Hereditary Autoinflammatory Diseases/metabolism , Humans , Lipoylation/drug effects , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/analysis , Small Molecule Libraries/metabolism
6.
Nat Commun ; 8(1): 427, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874664

ABSTRACT

Mammalian cells use cytosolic nucleic acid receptors to detect pathogens and other stress signals. In innate immune cells the presence of cytosolic DNA is sensed by the cGAS-STING signalling pathway, which initiates a gene expression programme linked to cellular activation and cytokine production. Whether the outcome of the STING response varies between distinct cell types remains largely unknown. Here we show that T cells exhibit an intensified STING response, which leads to the expression of a distinct set of genes and results in the induction of apoptosis. Of note, this proapoptotic STING response is still functional in cancerous T cells and delivery of small molecule STING agonists prevents in vivo growth of T-cell-derived tumours independent of its adjuvant activity. Our results demonstrate how the magnitude of STING signalling can shape distinct effector responses, which may permit for cell type-adjusted behaviours towards endogenous or exogenous insults.The cGAS/STING signalling pathway is responsible for sensing intracellular DNA and activating downstream inflammatory genes. Here the authors show mouse primary T cells and T leukaemia are hyperresponsive to STING agonist, and this strong STING signalling is associated with apoptosis induction.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Signal Transduction , Animals , Interferon Regulatory Factor-3/metabolism , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology , Mice, Inbred C57BL , Protein Binding , T-Lymphocytes/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
7.
Immunity ; 43(4): 727-38, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26431948

ABSTRACT

Whether interleukin-17A (IL-17A) has pathogenic and/or protective roles in the gut mucosa is controversial and few studies have analyzed specific cell populations for protective functions within the inflamed colonic tissue. Here we have provided evidence for IL-17A-dependent regulation of the tight junction protein occludin during epithelial injury that limits excessive permeability and maintains barrier integrity. Analysis of epithelial cells showed that in the absence of signaling via the IL-17 receptor adaptor protein Act-1, the protective effect of IL-17A was abrogated and inflammation was enhanced. We have demonstrated that after acute intestinal injury, IL-23R(+) γδ T cells in the colonic lamina propria were the primary producers of early, gut-protective IL-17A, and this production of IL-17A was IL-23 independent, leaving protective IL-17 intact in the absence of IL-23. These results suggest that IL-17-producing γδ T cells are important for the maintenance and protection of epithelial barriers in the intestinal mucosa.


Subject(s)
Colitis/physiopathology , Interleukin-17/physiology , Interleukin-23/physiology , Intestinal Mucosa/physiopathology , Acute Disease , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Line, Tumor , Cell Polarity , Colitis/chemically induced , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelium/physiopathology , Homeodomain Proteins/physiology , Humans , Interleukin-17/deficiency , Interleukin-17/pharmacology , Lymphocyte Subsets/metabolism , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Occludin/metabolism , Permeability , Protein Transport , Receptors, Antigen, T-Cell, gamma-delta/analysis , Recombinant Proteins/pharmacology , Tight Junctions/physiology , Tumor Necrosis Factor-alpha/pharmacology
8.
Cell Host Microbe ; 18(3): 263-5, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26355209

ABSTRACT

The DNA sensor, cyclic GMP-AMP synthase (cGAS), is essential for the detection of viral infection. In a recent issue of Science, two studies, Bridgeman et al. (2015) and Gentili et al. (2015), report a novel mechanism for propagating an antiviral signal between cells, based on the transfer of the cGAS enzymatic product, cyclic GMP-AMP (cGAMP), in viral particles.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/metabolism , Herpes Simplex/immunology , Herpesviridae Infections/immunology , Herpesvirus 1, Human/metabolism , Immunity, Innate/immunology , Interferon-beta/immunology , Muromegalovirus/metabolism , Nucleotides, Cyclic/metabolism , Second Messenger Systems , Vaccinia virus/metabolism , Vaccinia/immunology , Virion/metabolism , Animals , Humans
9.
Gastroenterology ; 149(7): 1860-1871.e8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26344057

ABSTRACT

BACKGROUND & AIMS: Single immunoglobulin and toll-interleukin 1 receptor (SIGIRR), a negative regulator of the Toll-like and interleukin-1 receptor (IL-1R) signaling pathways, controls intestinal inflammation and suppresses colon tumorigenesis in mice. However, the importance of SIGIRR in human colorectal cancer development has not been determined. We investigated the role of SIGIRR in development of human colorectal cancer. METHODS: We performed RNA sequence analyses of pairs of colon tumor and nontumor tissues, each collected from 68 patients. Immunoblot and immunofluorescence analyses were used to determine levels of SIGIRR protein in primary human colonic epithelial cells, tumor tissues, and colon cancer cell lines. We expressed SIGIRR and mutant forms of the protein in Vaco cell lines. We created and analyzed mice that expressed full-length (control) or a mutant form of Sigirr (encoding SIGIRR(N86/102S), which is not glycosylated) specifically in the intestinal epithelium. Some mice were given azoxymethane (AOM) and dextran sulfate sodium to induce colitis-associated cancer. Intestinal tissues were collected and analyzed by immunohistochemical and gene expression profile analyses. RESULTS: RNA sequence analyses revealed increased expression of a SIGIRR mRNA isoform, SIGIRR(ΔE8), in colorectal cancer tissues compared to paired nontumor tissues. SIGIRR(ΔE8) is not modified by complex glycans and is therefore retained in the cytoplasm-it cannot localize to the cell membrane or reduce IL1R signaling. SIGIRR(ΔE8) interacts with and has a dominant-negative effect on SIGIRR, reducing its glycosylation, localization to the cell surface, and function. Most SIGIRR detected in human colon cancer tissues was cytoplasmic, whereas in nontumor tissues it was found at the cell membrane. Mice that expressed SIGIRR(N86/102S) developed more inflammation and formed larger tumors after administration of azoxymethane and dextran sulfate sodium than control mice; colon tissues from these mutant mice expressed higher levels of the inflammatory cytokines IL-17A and IL-6 had activation of the transcription factors STAT3 and NFκB. SIGIRR(N86/102S) expressed in colons of mice did not localize to the epithelial cell surface. CONCLUSION: Levels of SIGIRR are lower in human colorectal tumors, compared with nontumor tissues; tumors contain the dominant-negative isoform SIGIRR(ΔE8). This mutant protein blocks localization of full-length SIGIRR to the surface of colon epithelial cells and its ability to downregulate IL1R signaling. Expression of SIGIRR(N86/102S) in the colonic epithelium of mice increases expression of inflammatory cytokines and formation and size of colitis-associated tumors.


Subject(s)
Colitis/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Genes, Dominant , Intestinal Mucosa/metabolism , Receptors, Interleukin-1/metabolism , Animals , Azoxymethane , Cell Membrane/metabolism , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytosol/metabolism , Dextran Sulfate , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Glycosylation , HeLa Cells , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , Protein Processing, Post-Translational , RNA, Messenger/metabolism , Receptors, Interleukin-1/genetics , Signal Transduction , Tissue Culture Techniques , Transfection
10.
Cell Host Microbe ; 17(6): 799-810, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26048138

ABSTRACT

Cytosolic detection of microbial products is essential for the initiation of an innate immune response against intracellular pathogens such as Mycobacterium tuberculosis (Mtb). During Mtb infection of macrophages, activation of cytosolic surveillance pathways is dependent on the mycobacterial ESX-1 secretion system and leads to type I interferon (IFN) and interleukin-1ß (IL-1ß) production. Whereas the inflammasome regulates IL-1ß secretion, the receptor(s) responsible for the activation of type I IFNs has remained elusive. We demonstrate that the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) is essential for initiating an IFN response to Mtb infection. cGAS associates with Mtb DNA in the cytosol to stimulate cyclic GAMP (cGAMP) synthesis. Notably, activation of cGAS-dependent cytosolic host responses can be uncoupled from inflammasome activation by modulating the secretion of ESX-1 substrates. Our findings identify cGAS as an innate sensor of Mtb and provide insight into how ESX-1 controls the activation of specific intracellular recognition pathways.


Subject(s)
DNA, Bacterial/metabolism , Host-Pathogen Interactions/immunology , Interferon Type I/metabolism , Mycobacterium tuberculosis/genetics , Nucleotidyltransferases/metabolism , Tuberculosis/microbiology , Animals , Female , Humans , Male
11.
Nat Commun ; 5: 4977, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25266676

ABSTRACT

The inflammasomes are multiprotein complexes that activate caspase-1 in response to infections and stress, resulting in the secretion of pro-inflammatory cytokines. Here we report that IκB kinase α (IKKα) is a critical negative regulator of apoptosis-associated specklike protein containing a C-terminal caspase-activation-andrecruitment (CARD) domain (ASC)-dependent inflammasomes. IKKα controls the inflammasome at the level of the adaptor ASC, which interacts with IKKα in the nucleus of resting macrophages in an IKKα kinase-dependent manner. Loss of IKKα kinase activity results in inflammasome hyperactivation. Mechanistically, the downstream nuclear effector IKK-related kinase (IKKi) facilitates translocation of ASC from the nucleus to the perinuclear area during inflammasome activation. ASC remains under the control of IKKα in the perinuclear area following translocation of the ASC/IKKα complex. Signal 2 of NLRP3 activation leads to inhibition of IKKα kinase activity through the recruitment of PP2A, allowing ASC to participate in NLRP3 inflammasome assembly. Taken together, these findings reveal a IKKi-IKKα-ASC axis that serves as a common regulatory mechanism for ASC-dependent inflammasomes.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Down-Regulation , I-kappa B Kinase/metabolism , Inflammasomes/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Female , I-kappa B Kinase/genetics , Inflammasomes/genetics , Macrophages/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Transport
12.
PLoS Pathog ; 9(8): e1003539, 2013.
Article in English | MEDLINE | ID: mdl-23950714

ABSTRACT

Enteric bacterial pathogens such as enterohemorrhagic E. coli (EHEC) and Salmonella Typhimurium target the intestinal epithelial cells (IEC) lining the mammalian gastrointestinal tract. Despite expressing innate Toll-like receptors (TLRs), IEC are innately hypo-responsive to most bacterial products. This is thought to prevent maladaptive inflammatory responses against commensal bacteria, but it also limits antimicrobial responses by IEC to invading bacterial pathogens, potentially increasing host susceptibility to infection. One reason for the innate hypo-responsiveness of IEC is their expression of Single Ig IL-1 Related Receptor (SIGIRR), a negative regulator of interleukin (IL)-1 and TLR signaling. To address whether SIGIRR expression and the innate hypo-responsiveness of IEC impacts on enteric host defense, Sigirr deficient (-/-) mice were infected with the EHEC related pathogen Citrobacter rodentium. Sigirr -/- mice responded with accelerated IEC proliferation and strong pro-inflammatory and antimicrobial responses but surprisingly, Sigirr -/- mice proved dramatically more susceptible to infection than wildtype mice. Through haematopoietic transplantation studies, it was determined that SIGIRR expression by non-haematopoietic cells (putative IEC) regulated these responses. Moreover, the exaggerated responses were found to be primarily dependent on IL-1R signaling. Whilst exploring the basis for their susceptibility, Sigirr -/- mice were found to be unusually susceptible to intestinal Salmonella Typhimurium colonization, developing enterocolitis without the typical requirement for antibiotic based removal of competing commensal microbes. Strikingly, the exaggerated antimicrobial responses seen in Sigirr -/- mice were found to cause a rapid and dramatic loss of commensal microbes from the infected intestine. This depletion appears to reduce the ability of the microbiota to compete for space and nutrients (colonization resistance) with the invading pathogens, leaving the intestine highly susceptible to pathogen colonization. Thus, SIGIRR expression by IEC reflects a strategy that sacrifices maximal innate responsiveness by IEC in order to promote commensal microbe based colonization resistance against bacterial pathogens.


Subject(s)
Enterobacteriaceae Infections/immunology , Enterobacteriaceae/immunology , Immunity, Innate , Receptors, Interleukin-1/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , Enterobacteriaceae/genetics , Enterobacteriaceae Infections/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Mice , Mice, Knockout , Receptors, Interleukin-1/genetics , Signal Transduction/genetics , Toll-Like Receptors/genetics
13.
Gut ; 62(2): 209-19, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22345661

ABSTRACT

OBJECTIVE: Intestinal microflora and inflammatory cell infiltrates play critical roles in the pathogenesis of acute colitis. Ceruloplasmin is an acute-phase plasma protein produced by hepatocytes and activated macrophages, and has ferroxidase with bactericidal activities. The goal is to understand the role of ceruloplasmin in colitis progression in a genetically modified murine model. DESIGN: Experimental colitis was induced in ceruloplasmin null (Cp(-/-)) and wild-type (WT) mice by dextran sulphate sodium administration. The role of ceruloplasmin was further evaluated by transplantation of WT macrophages into Cp(-/-) mice. RESULTS: Cp(-/-) mice rapidly lost weight and were moribund by day 14, while WT mice survived at least 30 days. Colon culture supernatants from Cp(-/-) mice exhibited elevated levels of TNFα, KC and MCP-1, indicative of increased inflammation and neutrophil and macrophage infiltration. Elevated leucocytes and severe histopathology were observed in Cp(-/-) mice. Elevated protein carbonyl content was detected in colons from Cp(-/-) mice suggesting ceruloplasmin antioxidant activity might contribute to its protective function. Unexpectedly, intraperitoneal administration of human ceruloplasmin into Cp(-/-) mice did not afford protection. Bone marrow transplantation from WT mice or injection of isolated peripheral blood monocytes markedly reduced severity of colitis and morbidity in Cp(-/-) mice. CONCLUSION: Macrophage-derived ceruloplasmin contributes importantly to protection against inflammation and tissue injury in acute and chronic experimental colitis. The findings suggest that defects in ceruloplasmin expression or processing may influence the onset or progression of inflammatory bowel disease in patients.


Subject(s)
Ceruloplasmin/physiology , Colitis/prevention & control , Macrophages, Peritoneal/metabolism , Animals , Bone Marrow Transplantation , Chemokines/metabolism , Colitis/chemically induced , Colitis/metabolism , DNA Primers/chemistry , Dextran Sulfate , Disease Progression , Female , Mice , Mice, Inbred C57BL , Protein Carbonylation , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
14.
Immunity ; 37(5): 800-12, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23142783

ABSTRACT

Interleukin-1 (IL-1)-induced activation of the mTOR kinase pathway has major influences on Th17 cell survival, proliferation, and effector function. Via biochemical and genetic approaches, the kinases IKKi and GSK3α were identified as the critical intermediate signaling components for IL-1-induced AKT activation, which in turn activated mTOR. Although insulin-induced AKT activation is known to phosphorylate and inactivate GSK3α and GSK3ß, we found that GSK3α but not GSK3ß formed a constitutive complex to phosphorylate and suppress AKT activation, showing that a reverse action from GSK to AKT can take place. Upon IL-1 stimulation, IKKi was activated to mediate GSK3α phosphorylation at S21, thereby inactivating GSK3α to promote IL-1-induced AKT-mTOR activation. Thus, IKKi has a critical role in Th17 cell maintenance and/or proliferation through the GSK-AKT-mTOR pathway, implicating the potential of IKKi as a therapeutic target.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , I-kappa B Kinase/metabolism , Interleukin-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Th17 Cells/metabolism , Animals , Cell Growth Processes/physiology , Enzyme Activation , Glycogen Synthase Kinase 3/immunology , Glycogen Synthase Kinase 3 beta , Insulin/immunology , Insulin/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , TOR Serine-Threonine Kinases/immunology , Th17 Cells/cytology , Th17 Cells/enzymology , Th17 Cells/immunology
15.
J Immunol ; 189(1): 33-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22649194

ABSTRACT

The effector T cell subset, Th17, plays a significant role in the pathogenesis of multiple sclerosis and of other autoimmune diseases. The signature cytokine, IL-17, engages the IL-17R and recruits the E3-ligase NF-κB activator 1 (Act1) upon stimulation. In this study, we examined the role of TNFR-associated factor (TRAF)4 in IL-17 signaling and Th17-mediated autoimmune encephalomyelitis. Primary cells from TRAF4-deficient mice displayed markedly enhanced IL-17-activated signaling pathways and induction of chemokine mRNA. Adoptive transfer of MOG35-55 specific wild-type Th17 cells into TRAF4-deficient recipient mice induced an earlier onset of disease. Mechanistically, we found that TRAF4 and TRAF6 used the same TRAF binding sites on Act1, allowing the competition of TRAF4 with TRAF6 for the interaction with Act1. Taken together, the results of this study reveal the necessity of a unique role of TRAF4 in restricting the effects of IL-17 signaling and Th17-mediated disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-17/antagonists & inhibitors , Interleukin-17/physiology , Signal Transduction/immunology , TNF Receptor-Associated Factor 4/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , HEK293 Cells , HeLa Cells , Humans , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , Signal Transduction/genetics , TNF Receptor-Associated Factor 4/deficiency , TNF Receptor-Associated Factor 4/genetics , Th17 Cells/immunology , Th17 Cells/pathology
16.
Nat Immunol ; 12(9): 844-52, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822257

ABSTRACT

Interleukin 17 (IL-17) is critical in the pathogenesis of inflammatory and autoimmune diseases. Here we report that Act1, the key adaptor for the IL-17 receptor (IL-7R), formed a complex with the inducible kinase IKKi after stimulation with IL-17. Through the use of IKKi-deficient mice, we found that IKKi was required for IL-17-induced expression of genes encoding inflammatory molecules in primary airway epithelial cells, neutrophilia and pulmonary inflammation. IKKi deficiency abolished IL-17-induced formation of the complex of Act1 and the adaptors TRAF2 and TRAF5, activation of mitogen-activated protein kinases (MAPKs) and mRNA stability, whereas the Act1-TRAF6-transcription factor NF-κB axis was retained. IKKi was required for IL-17-induced phosphorylation of Act1 on Ser311, adjacent to a putative TRAF-binding motif. Substitution of the serine at position 311 with alanine impaired the IL-17-mediated Act1-TRAF2-TRAF5 interaction and gene expression. Thus, IKKi is a kinase newly identified as modulating IL-17 signaling through its effect on Act1 phosphorylation and consequent function.


Subject(s)
Adaptor Proteins, Signal Transducing , Chemokine CXCL1/immunology , I-kappa B Kinase , Neutrophils/immunology , Pneumonia/immunology , Signal Transduction/immunology , Th17 Cells/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression Regulation , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-17/pharmacology , Lung , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/immunology , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/metabolism , Phosphorylation , Pneumonia/genetics , Pneumonia/metabolism , RNA Stability/drug effects , RNA, Messenger , Receptors, Interleukin-17/immunology , TNF Receptor-Associated Factor 5/immunology , TNF Receptor-Associated Factor 5/metabolism , Th17 Cells/metabolism
17.
Gastroenterology ; 139(2): 574-85, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20416302

ABSTRACT

BACKGROUND & AIMS: Commensal bacteria can activate signaling by the Toll-like and interleukin-1 receptors (TLR and IL-1R) to mediate pathogenesis of inflammatory bowel diseases and colitis-associated cancer. We investigated the role of the single immunoglobulin IL-1 receptor-related (SIGIRR) molecule, a negative regulator of TLR and IL-1R signaling, as a tumor suppressor to determine whether SIGIRR controls cell-cycle progression, genetic instability, and colon tumor initiation by modulating commensal TLR signaling in the gastrointestinal tract. METHODS: We analyzed adenomatous polyposis coli (Apc)min/+/Sigirr-/- mice for polyps, microadenomas, and anaphase bridge index. Commensal bacteria were depleted from mice with antibiotics. Akt, mammalian target of rapamycin (mTOR), and beta-catenin pathways were examined by immunoblotting and immunohistochemistry. Loss of heterozygosity of Apc and expression of cytokines and proinflammatory mediators were measured by nonquantitative or quantitative polymerase chain reaction. RESULTS: Apcmin/+/Sigirr-/- mice had increased loss of heterozygosity of Apc and microadenoma formation, resulting in spontaneous colonic polyposis, compared with Apcmin/+/Sigirr+/+ mice. The increased colonic tumorigenesis that occurred in the Apcmin/+/Sigirr-/- mice depended on the presence of commensal bacteria in the gastrointestinal tract. Cell proliferation and chromosomal instability increased in colon crypt cells of the Apcmin/+/Sigirr-/- mice. Akt, mTOR, and their substrates were hyperactivated in colon epithelium of Apcmin/+/Sigirr-/- mice in response to TLR or IL-1R ligands. Inhibition of the mTOR pathway by rapamycin reduced formation of microadenomas and polyps in the Apcmin/+/Sigirr-/- mice. CONCLUSIONS: SIGIRR acts as a tumor suppressor in the colon by inhibiting TLR-induced, mTOR-mediated cell-cycle progression and genetic instability.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/prevention & control , Colon/metabolism , Genes, APC , Genes, Tumor Suppressor , Receptors, Interleukin-1/deficiency , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Animals , Blotting, Western , Cell Proliferation , Chromosomal Instability , Colon/drug effects , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Immunohistochemistry , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-1/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Toll-Like Receptors/metabolism , Tumor Burden , beta Catenin/metabolism
18.
Immunity ; 32(1): 54-66, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20060329

ABSTRACT

Interleukin-1 (IL-1)-mediated signaling in T cells is essential for T helper 17 (Th17) cell differentiation. We showed here that SIGIRR, a negative regulator of IL-1 receptor and Toll-like receptor signaling, was induced during Th17 cell lineage commitment and governed Th17 cell differentiation and expansion through its inhibitory effects on IL-1 signaling. The absence of SIGIRR in T cells resulted in increased Th17 cell polarization in vivo upon myelin oligodendrocyte glycoprotein (MOG(35-55)) peptide immunization. Recombinant IL-1 promoted a marked increase in the proliferation of SIGIRR-deficient T cells under an in vitro Th17 cell-polarization condition. Importantly, we detected increased IL-1-induced phosphorylation of JNK and mTOR kinase in SIGIRR-deficient Th17 cells compared to wild-type Th17 cells. IL-1-induced proliferation was abolished in mTOR-deficient Th17 cells, indicating the essential role of mTOR activation. Our results demonstrate an important mechanism by which SIGIRR controls Th17 cell expansion and effector function through the IL-1-induced mTOR signaling pathway.


Subject(s)
Cell Differentiation/immunology , Interleukin-17/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Receptors, Interleukin-1/immunology , T-Lymphocytes, Helper-Inducer/cytology , Animals , Cell Lineage/immunology , Cell Proliferation , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Immunoprecipitation , Interleukin-17/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases , Transfection
19.
J Immunol ; 183(1): 568-77, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542468

ABSTRACT

Both IL-23- and IL-1-mediated signaling pathways play important roles in Th17 cell differentiation, cytokine production, and autoimmune diseases. The IL-1R-associated kinase 4 (IRAK4) is critical for IL-1/TLR signaling. We show here that inactivation of IRAK4 kinase in mice (IRAK4 KI) results in significant resistance to experimental autoimmune encephalomyelitis due to a reduction in infiltrating inflammatory cells into the CNS and reduced Ag-specific CD4(+) T cell-mediated IL-17 production. Adoptive transfer of myelin oligodendrocyte glycoprotein 35-55-specific IRAK4 KI Th17 cells failed to induce experimental autoimmune encephalomyelitis in either wild-type or IRAK4 KI recipient mice, indicating the lack of autoantigen-specific Th17 cell activities in the absence of IRAK4 kinase activity. Furthermore, the absence of IRAK4 kinase activity blocked induction of IL-23R expression, STAT3 activation by IL-23, and Th17 cytokine expression in differentiated Th17 cells. Importantly, blockade of IL-1 signaling by IL-1RA inhibited Th17 differentiation and IL-23-induced cytokine expression in differentiated Th17 cells. The results of these studies demonstrate that IL-1-mediated IRAK4 kinase activity in T cells is essential for induction of IL-23R expression, Th17 differentiation, and autoimmune disease.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/physiology , Interleukin-17/physiology , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cell Migration Inhibition/genetics , Cell Migration Inhibition/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme Activation/genetics , Enzyme Activation/immunology , Female , Gene Knock-In Techniques , Glycoproteins/administration & dosage , Glycoproteins/antagonists & inhibitors , Immunity, Innate/genetics , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Mice , Molecular Sequence Data , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Peptide Fragments/antagonists & inhibitors , Signal Transduction/genetics , Signal Transduction/immunology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes, Helper-Inducer/pathology
20.
J Immunol ; 182(5): 2601-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234154

ABSTRACT

A novel cytokine IL-33, an IL-1 family member, signals via ST2 receptor and promotes Th2 responses, through the activation of NF-kappaB and MAP kinases. Previous studies reported that single Ig IL-1R-related molecule (SIGIRR)/Toll IL-1R8 acts as negative regulator for TLR-IL-1R-mediated signaling. We now found that SIGIRR formed a complex with ST2 upon IL-33 stimulation and specifically inhibited IL-33/ST2-mediated signaling in cell culture model. Furthermore, IL-33-induced Th2 response was enhanced in SIGIRR-deficient mice compared with that in wild-type control mice, suggesting a negative regulatory role of SIGIRR in IL-33/ST2 signaling in vivo. Similar to ST2, SIGIRR was highly expressed in in vitro polarized Th2 cells, but not Th1 cells. SIGIRR-deficient Th2 cells produce higher levels of Th2 cytokines, including IL-5, IL-4, and IL-13, than that in wild-type cells. Moreover, SIGIRR-deficient mice developed stronger Th2 immune response in OVA-challenged asthma model. Taken together, our results suggest that SIGIRR plays an important role in the regulation of Th2 response in vivo, possibly through its impact on IL-33-ST2-mediated signaling.


Subject(s)
Receptors, Interleukin-1/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Cell Line , Cells, Cultured , Down-Regulation/immunology , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/administration & dosage , Interleukins/antagonists & inhibitors , Interleukins/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/immunology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...