Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
Add more filters

Publication year range
1.
Cell ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38878777

ABSTRACT

NLRs constitute a large, highly conserved family of cytosolic pattern recognition receptors that are central to health and disease, making them key therapeutic targets. NLRC5 is an enigmatic NLR with mutations associated with inflammatory and infectious diseases, but little is known about its function as an innate immune sensor and cell death regulator. Therefore, we screened for NLRC5's role in response to infections, PAMPs, DAMPs, and cytokines. We identified that NLRC5 acts as an innate immune sensor to drive inflammatory cell death, PANoptosis, in response to specific ligands, including PAMP/heme and heme/cytokine combinations. NLRC5 interacted with NLRP12 and PANoptosome components to form a cell death complex, suggesting an NLR network forms similar to those in plants. Mechanistically, TLR signaling and NAD+ levels regulated NLRC5 expression and ROS production to control cell death. Furthermore, NLRC5-deficient mice were protected in hemolytic and inflammatory models, suggesting that NLRC5 could be a potential therapeutic target.

2.
Cell ; 186(13): 2783-2801.e20, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37267949

ABSTRACT

Cytosolic innate immune sensors are critical for host defense and form complexes, such as inflammasomes and PANoptosomes, that induce inflammatory cell death. The sensor NLRP12 is associated with infectious and inflammatory diseases, but its activating triggers and roles in cell death and inflammation remain unclear. Here, we discovered that NLRP12 drives inflammasome and PANoptosome activation, cell death, and inflammation in response to heme plus PAMPs or TNF. TLR2/4-mediated signaling through IRF1 induced Nlrp12 expression, which led to inflammasome formation to induce maturation of IL-1ß and IL-18. The inflammasome also served as an integral component of a larger NLRP12-PANoptosome that drove inflammatory cell death through caspase-8/RIPK3. Deletion of Nlrp12 protected mice from acute kidney injury and lethality in a hemolytic model. Overall, we identified NLRP12 as an essential cytosolic sensor for heme plus PAMPs-mediated PANoptosis, inflammation, and pathology, suggesting that NLRP12 and molecules in this pathway are potential drug targets for hemolytic and inflammatory diseases.


Subject(s)
Inflammasomes , Pathogen-Associated Molecular Pattern Molecules , Animals , Mice , Inflammasomes/metabolism , Heme , Inflammation , Pyroptosis , Intracellular Signaling Peptides and Proteins
3.
Cell ; 184(1): 149-168.e17, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33278357

ABSTRACT

COVID-19 is characterized by excessive production of pro-inflammatory cytokines and acute lung damage associated with patient mortality. While multiple inflammatory cytokines are produced by innate immune cells during SARS-CoV-2 infection, we found that only the combination of TNF-α and IFN-γ induced inflammatory cell death characterized by inflammatory cell death, PANoptosis. Mechanistically, TNF-α and IFN-γ co-treatment activated the JAK/STAT1/IRF1 axis, inducing nitric oxide production and driving caspase-8/FADD-mediated PANoptosis. TNF-α and IFN-γ caused a lethal cytokine shock in mice that mirrors the tissue damage and inflammation of COVID-19, and inhibiting PANoptosis protected mice from this pathology and death. Furthermore, treating with neutralizing antibodies against TNF-α and IFN-γ protected mice from mortality during SARS-CoV-2 infection, sepsis, hemophagocytic lymphohistiocytosis, and cytokine shock. Collectively, our findings suggest that blocking the cytokine-mediated inflammatory cell death signaling pathway identified here may benefit patients with COVID-19 or other infectious and autoinflammatory diseases by limiting tissue damage/inflammation.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Interferon-gamma/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Cell Death , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/immunology , Inflammation/pathology , Lymphohistiocytosis, Hemophagocytic/chemically induced , Male , Mice , Mice, Transgenic , THP-1 Cells
4.
Nat Immunol ; 24(10): 1735-1747, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37679549

ABSTRACT

Neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by innate immune-mediated inflammation, but functional and mechanistic effects of the adaptive immune system remain unclear. Here we identify brain-resident CD8+ T cells that coexpress CXCR6 and PD-1 and are in proximity to plaque-associated microglia in human and mouse AD brains. We also establish that CD8+ T cells restrict AD pathologies, including ß-amyloid deposition and cognitive decline. Ligand-receptor interaction analysis identifies CXCL16-CXCR6 intercellular communication between microglia and CD8+ T cells. Further, Cxcr6 deficiency impairs accumulation, tissue residency programming and clonal expansion of brain PD-1+CD8+ T cells. Ablation of Cxcr6 or CD8+ T cells ultimately increases proinflammatory cytokine production from microglia, with CXCR6 orchestrating brain CD8+ T cell-microglia colocalization. Collectively, our study reveals protective roles for brain CD8+ T cells and CXCR6 in mouse AD pathogenesis and highlights that microenvironment-specific, intercellular communication orchestrates tissue homeostasis and protection from neuroinflammation.

5.
Cell ; 181(3): 674-687.e13, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32298652

ABSTRACT

Caspases regulate cell death, immune responses, and homeostasis. Caspase-6 is categorized as an executioner caspase but shows key differences from the other executioners. Overall, little is known about the functions of caspase-6 in biological processes apart from apoptosis. Here, we show that caspase-6 mediates innate immunity and inflammasome activation. Furthermore, we demonstrate that caspase-6 promotes the activation of programmed cell death pathways including pyroptosis, apoptosis, and necroptosis (PANoptosis) and plays an essential role in host defense against influenza A virus (IAV) infection. In addition, caspase-6 promoted the differentiation of alternatively activated macrophages (AAMs). Caspase-6 facilitated the RIP homotypic interaction motif (RHIM)-dependent binding of RIPK3 to ZBP1 via its interaction with RIPK3. Altogether, our findings reveal a vital role for caspase-6 in facilitating ZBP1-mediated inflammasome activation, cell death, and host defense during IAV infection, opening additional avenues for treatment of infectious and autoinflammatory diseases and cancer.


Subject(s)
Caspase 6/immunology , Caspase 6/metabolism , Inflammasomes/immunology , Animals , Apoptosis/immunology , Cell Death/immunology , Immunity, Innate , Inflammasomes/metabolism , Inflammasomes/physiology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Necroptosis/immunology , Protein Binding , Pyroptosis/immunology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
6.
Nat Immunol ; 22(7): 829-838, 2021 07.
Article in English | MEDLINE | ID: mdl-33963333

ABSTRACT

The innate immune response is critical for recognizing and controlling infections through the release of cytokines and chemokines. However, severe pathology during some infections, including SARS-CoV-2, is driven by hyperactive cytokine release, or a cytokine storm. The innate sensors that activate production of proinflammatory cytokines and chemokines during COVID-19 remain poorly characterized. In the present study, we show that both TLR2 and MYD88 expression were associated with COVID-19 disease severity. Mechanistically, TLR2 and Myd88 were required for ß-coronavirus-induced inflammatory responses, and TLR2-dependent signaling induced the production of proinflammatory cytokines during coronavirus infection independent of viral entry. TLR2 sensed the SARS-CoV-2 envelope protein as its ligand. In addition, blocking TLR2 signaling in vivo provided protection against the pathogenesis of SARS-CoV-2 infection. Overall, our study provides a critical understanding of the molecular mechanism of ß-coronavirus sensing and inflammatory cytokine production, which opens new avenues for therapeutic strategies to counteract the ongoing COVID-19 pandemic.


Subject(s)
COVID-19/immunology , Coronavirus Envelope Proteins/metabolism , Cytokine Release Syndrome/immunology , SARS-CoV-2/immunology , Toll-Like Receptor 2/metabolism , Animals , COVID-19/complications , COVID-19/diagnosis , COVID-19/virology , Chlorocebus aethiops , Cytokine Release Syndrome/diagnosis , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunity, Innate/drug effects , Leukocytes, Mononuclear , Macrophages , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Primary Cell Culture , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Vero Cells , COVID-19 Drug Treatment
7.
Cell ; 162(1): 45-58, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095253

ABSTRACT

Colorectal cancer is a leading cause of cancer-related deaths. Mutations in the innate immune sensor AIM2 are frequently identified in patients with colorectal cancer, but how AIM2 modulates colonic tumorigenesis is unknown. Here, we found that Aim2-deficient mice were hypersusceptible to colonic tumor development. Production of inflammasome-associated cytokines and other inflammatory mediators was largely intact in Aim2-deficient mice; however, intestinal stem cells were prone to uncontrolled proliferation. Aberrant Wnt signaling expanded a population of tumor-initiating stem cells in the absence of AIM2. Susceptibility of Aim2-deficient mice to colorectal tumorigenesis was enhanced by a dysbiotic gut microbiota, which was reduced by reciprocal exchange of gut microbiota with healthy wild-type mice. These findings uncover a synergy between a specific host genetic factor and gut microbiota in determining the susceptibility to colorectal cancer. Therapeutic modulation of AIM2 expression and microbiota has the potential to prevent colorectal cancer.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Stem Cells/pathology , Animals , Azoxymethane , Colitis/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dextran Sulfate , Enterocytes/pathology , Gastrointestinal Tract/microbiology , Inflammasomes/metabolism , Mice , Mutation , Stem Cells/metabolism
8.
Nature ; 628(8009): 835-843, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600381

ABSTRACT

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Subject(s)
Lung Injury , Necroptosis , Orthomyxoviridae Infections , Protein Kinase Inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Female , Humans , Male , Mice , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/metabolism , Influenza A virus/classification , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung Injury/complications , Lung Injury/pathology , Lung Injury/prevention & control , Lung Injury/virology , Mice, Inbred C57BL , Necroptosis/drug effects , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/prevention & control , Respiratory Distress Syndrome/virology
9.
Nat Immunol ; 17(3): 277-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808230

ABSTRACT

Regulatory T (Treg) cells respond to immune and inflammatory signals to mediate immunosuppression, but how the functional integrity of Treg cells is maintained under activating environments is unclear. Here we show that autophagy is active in Treg cells and supports their lineage stability and survival fitness. Treg cell-specific deletion of Atg7 or Atg5, two essential genes in autophagy, leads to loss of Treg cells, greater tumor resistance and development of inflammatory disorders. Atg7-deficient Treg cells show increased apoptosis and readily lose expression of the transcription factor Foxp3, especially after activation. Mechanistically, autophagy deficiency upregulates metabolic regulators mTORC1 and c-Myc and glycolysis, which contribute to defective Treg function. Therefore, autophagy couples environmental signals and metabolic homeostasis to protect lineage and survival integrity of Treg cells in activating contexts.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Forkhead Transcription Factors/genetics , Microtubule-Associated Proteins/genetics , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Adenocarcinoma/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , Autophagy/immunology , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Line, Tumor , Colonic Neoplasms/immunology , DNA Methylation , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Glycolysis , Homeostasis , Immunoblotting , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Neoplasm Transplantation , Real-Time Polymerase Chain Reaction , Up-Regulation
10.
Immunity ; 51(6): 1012-1027.e7, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31668641

ABSTRACT

Regulatory T (Treg) cells are critical mediators of immune tolerance whose activity depends upon T cell receptor (TCR) and mTORC1 kinase signaling, but the mechanisms that dictate functional activation of these pathways are incompletely understood. Here, we showed that amino acids license Treg cell function by priming and sustaining TCR-induced mTORC1 activity. mTORC1 activation was induced by amino acids, especially arginine and leucine, accompanied by the dynamic lysosomal localization of the mTOR and Tsc complexes. Rag and Rheb GTPases were central regulators of amino acid-dependent mTORC1 activation in effector Treg (eTreg) cells. Mice bearing RagA-RagB- or Rheb1-Rheb2-deficient Treg cells developed a fatal autoimmune disease and had reduced eTreg cell accumulation and function. RagA-RagB regulated mitochondrial and lysosomal fitness, while Rheb1-Rheb2 enforced eTreg cell suppressive gene signature. Together, these findings reveal a crucial requirement of amino acid signaling for licensing and sustaining mTORC1 activation and functional programming of Treg cells.


Subject(s)
Arginine/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Cycle , Cell Differentiation/physiology , Cell Line , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Ras Homolog Enriched in Brain Protein/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/cytology
11.
Nat Immunol ; 16(2): 178-87, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559258

ABSTRACT

The interplay between effector T cells and regulatory T cells (Treg cells) is crucial for adaptive immunity, but how Treg cells control diverse effector responses is elusive. We found that the phosphatase PTEN links Treg cell stability to repression of type 1 helper T cell (TH1 cell) and follicular helper T cell (TFH cell) responses. Depletion of PTEN in Treg cells resulted in excessive TFH cell and germinal center responses and spontaneous inflammatory disease. These defects were considerably blocked by deletion of interferon-γ, indicating coordinated control of TH1 and TFH responses. Mechanistically, PTEN maintained Treg cell stability and metabolic balance between glycolysis and mitochondrial fitness. Moreover, PTEN deficiency upregulates activity of the metabolic checkpoint kinase complex mTORC2 and the serine-threonine kinase Akt, and loss of this activity restores functioning of PTEN-deficient Treg cells. Our studies establish a PTEN-mTORC2 axis that maintains Treg cell stability and coordinates Treg cell-mediated control of effector responses.


Subject(s)
PTEN Phosphohydrolase/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , B-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Humans , Lymphocyte Activation , Mice , Repressor Proteins/metabolism , Signal Transduction , Th1 Cells/enzymology
12.
Nat Immunol ; 16(5): 467-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25774715

ABSTRACT

Inflammasomes are critical for mounting host defense against pathogens. The molecular mechanisms that control activation of the AIM2 inflammasome in response to different cytosolic pathogens remain unclear. Here we found that the transcription factor IRF1 was required for activation of the AIM2 inflammasome during infection with the Francisella tularensis subspecies novicida (F. novicida), whereas engagement of the AIM2 inflammasome by mouse cytomegalovirus (MCMV) or transfected double-stranded DNA did not require IRF1. Infection of F. novicida detected by the DNA sensor cGAS and its adaptor STING induced type I interferon-dependent expression of IRF1, which drove the expression of guanylate-binding proteins (GBPs); this led to intracellular killing of bacteria and DNA release. Our results reveal a specific requirement for IRF1 and GBPs in the liberation of DNA for sensing by AIM2 depending on the pathogen encountered by the cell.


Subject(s)
DNA-Binding Proteins/metabolism , Francisella tularensis/physiology , GTP-Binding Proteins/metabolism , Inflammasomes/metabolism , Interferon Regulatory Factor-1/metabolism , Tularemia/immunology , Animals , Bacteriolysis/genetics , Cells, Cultured , DNA/immunology , DNA, Bacterial/genetics , Gene Expression Regulation/genetics , Interferon Regulatory Factor-1/genetics , Interferon Type I/metabolism , Mice , Mice, Knockout , Nucleotidyltransferases/metabolism
13.
Immunity ; 49(3): 515-530.e5, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231985

ABSTRACT

Fungi represent a significant proportion of the gut microbiota. Aberrant immune responses to fungi are frequently observed in inflammatory bowel diseases (IBD) and colorectal cancer (CRC), and mutations in the fungal-sensing pathways are associated with the pathogenesis of IBD. Fungal recognition receptors trigger downstream signaling via the common adaptor protein CARD9 and the kinase SYK. Here we found that commensal gut fungi promoted inflammasome activation during AOM-DSS-induced colitis. Myeloid cell-specific deletion of Card9 or Syk reduced inflammasome activation and interleukin (IL)-18 maturation and increased susceptibility to colitis and CRC. IL-18 promoted epithelial barrier restitution and interferon-γ production by intestinal CD8+ T cells. Supplementation of IL-18 or transfer of wild-type myeloid cells reduced tumor burden in AOM-DSS-treated Card9-/- and Sykfl/flLysMCre/+ mice, whereas treatment with anti-fungal agents exacerbated colitis and CRC. CARD9 deletion changes the gut microbial landscape, suggesting that SYK-CARD9 signaling maintains a microbial ecology that promotes inflammasome activation and thereby restrains colitis and colon tumorigenesis.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Colitis/immunology , Colonic Neoplasms/immunology , Fungi/immunology , Gastrointestinal Microbiome/immunology , Inflammasomes/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/physiology , Myeloid Cells/physiology , Syk Kinase/metabolism , Animals , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Colitis/chemically induced , Disease Models, Animal , Humans , Interleukin-18/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Sodium Dodecyl Sulfate , Syk Kinase/genetics
14.
Immunity ; 49(3): 531-544.e6, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30170813

ABSTRACT

Compared to adults, infants suffer higher rates of hospitalization, severe clinical complications, and mortality due to influenza infection. We found that γδ T cells protected neonatal mice against mortality during influenza infection. γδ T cell deficiency did not alter viral clearance or interferon-γ production. Instead, neonatal influenza infection induced the accumulation of interleukin-17A (IL-17A)-producing γδ T cells, which was associated with IL-33 production by lung epithelial cells. Neonates lacking IL-17A-expressing γδ T cells or Il33 had higher mortality upon influenza infection. γδ T cells and IL-33 promoted lung infiltration of group 2 innate lymphoid cells and regulatory T cells, resulting in increased amphiregulin secretion and tissue repair. In influenza-infected children, IL-17A, IL-33, and amphiregulin expression were correlated, and increased IL-17A levels in nasal aspirates were associated with better clinical outcomes. Our results indicate that γδ T cells are required in influenza-infected neonates to initiate protective immunity and mediate lung homeostasis.


Subject(s)
Influenza A virus/physiology , Influenza, Human/immunology , Interleukin-17/metabolism , Lung/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Th2 Cells/immunology , Adult , Amphiregulin/metabolism , Animals , Cells, Cultured , Child , Humans , Immunity , Infant, Newborn , Interleukin-33/metabolism , Mice , Prognosis , Receptors, Antigen, T-Cell, gamma-delta/metabolism
15.
Immunity ; 49(5): 899-914.e6, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413360

ABSTRACT

Interleukin-2 (IL-2) and downstream transcription factor STAT5 are important for maintaining regulatory T (Treg) cell homeostasis and function. Treg cells can respond to low IL-2 levels, but the mechanisms of STAT5 activation during partial IL-2 deficiency remain uncertain. We identified the serine-threonine kinase Mst1 as a signal-dependent amplifier of IL-2-STAT5 activity in Treg cells. High Mst1 and Mst2 (Mst1-Mst2) activity in Treg cells was crucial to prevent tumor resistance and autoimmunity. Mechanistically, Mst1-Mst2 sensed IL-2 signals to promote the STAT5 activation necessary for Treg cell homeostasis and lineage stability and to maintain the highly suppressive phosphorylated-STAT5+ Treg cell subpopulation. Unbiased quantitative proteomics revealed association of Mst1 with the cytoskeletal DOCK8-LRCHs module. Mst1 deficiency limited Treg cell migration and access to IL-2 and activity of the small GTPase Rac, which mediated downstream STAT5 activation. Collectively, IL-2-STAT5 signaling depends upon Mst1-Mst2 functions to maintain a stable Treg cell pool and immune tolerance.


Subject(s)
Hepatocyte Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Lineage/genetics , Hepatocyte Growth Factor/genetics , Hippo Signaling Pathway , Interleukin-2/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Serine-Threonine Kinase 3 , T-Lymphocytes, Regulatory/immunology , rac GTP-Binding Proteins/metabolism
16.
Nature ; 591(7849): 306-311, 2021 03.
Article in English | MEDLINE | ID: mdl-33627871

ABSTRACT

Regulatory T cells (Treg cells) are essential for immune tolerance1, but also drive immunosuppression in the tumour microenvironment2. Therapeutic targeting of Treg cells in cancer will therefore require the identification of context-specific mechanisms that affect their function. Here we show that inhibiting lipid synthesis and metabolic signalling that are dependent on sterol-regulatory-element-binding proteins (SREBPs) in Treg cells unleashes effective antitumour immune responses without autoimmune toxicity. We find that the activity of SREBPs is upregulated in intratumoral Treg cells. Moreover, deletion of SREBP-cleavage-activating protein (SCAP)-a factor required for SREBP activity-in these cells inhibits tumour growth and boosts immunotherapy that is triggered by targeting the immune-checkpoint protein PD-1. These effects of SCAP deletion are associated with uncontrolled production of interferon-γ and impaired function of intratumoral Treg cells. Mechanistically, signalling through SCAP and SREBPs coordinates cellular programs for lipid synthesis and inhibitory receptor signalling in these cells. First, de novo fatty-acid synthesis mediated by fatty-acid synthase (FASN) contributes to functional maturation of Treg cells, and loss of FASN from Treg cells inhibits tumour growth. Second, Treg cells in tumours show enhanced expression of the PD-1 gene, through a process that depends on SREBP activity and signals via mevalonate metabolism to protein geranylgeranylation. Blocking PD-1 or SREBP signalling results in dysregulated activation of phosphatidylinositol-3-kinase in intratumoral Treg cells. Our findings show that metabolic reprogramming enforces the functional specialization of Treg cells in tumours, pointing to new ways of targeting these cells for cancer therapy.


Subject(s)
Lipid Metabolism , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Cholesterol/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Female , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mevalonic Acid/metabolism , Mice , Phosphatidylinositol 3-Kinase/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , Sterol Regulatory Element Binding Proteins/metabolism , T-Lymphocytes, Regulatory/enzymology , Up-Regulation
17.
Nature ; 600(7888): 308-313, 2021 12.
Article in English | MEDLINE | ID: mdl-34795452

ABSTRACT

Nutrients are emerging regulators of adaptive immunity1. Selective nutrients interplay with immunological signals to activate mechanistic target of rapamycin complex 1 (mTORC1), a key driver of cell metabolism2-4, but how these environmental signals are integrated for immune regulation remains unclear. Here we use genome-wide CRISPR screening combined with protein-protein interaction networks to identify regulatory modules that mediate immune receptor- and nutrient-dependent signalling to mTORC1 in mouse regulatory T (Treg) cells. SEC31A is identified to promote mTORC1 activation by interacting with the GATOR2 component SEC13 to protect it from SKP1-dependent proteasomal degradation. Accordingly, loss of SEC31A impairs T cell priming and Treg suppressive function in mice. In addition, the SWI/SNF complex restricts expression of the amino acid sensor CASTOR1, thereby enhancing mTORC1 activation. Moreover, we reveal that the CCDC101-associated SAGA complex is a potent inhibitor of mTORC1, which limits the expression of glucose and amino acid transporters and maintains T cell quiescence in vivo. Specific deletion of Ccdc101 in mouse Treg cells results in uncontrolled inflammation but improved antitumour immunity. Collectively, our results establish epigenetic and post-translational mechanisms that underpin how nutrient transporters, sensors and transducers interplay with immune signals for three-tiered regulation of mTORC1 activity and identify their pivotal roles in licensing T cell immunity and immune tolerance.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Nutrients , Protein Interaction Maps , T-Lymphocytes, Regulatory , Animals , Female , Male , Mice , Carrier Proteins/metabolism , CRISPR-Cas Systems/genetics , Forkhead Transcription Factors/metabolism , Genome/genetics , Homeostasis , Immune Tolerance , Inflammation/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/immunology , Nuclear Proteins/metabolism , Nutrients/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , S-Phase Kinase-Associated Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Trans-Activators/metabolism
18.
Mol Cell ; 74(4): 742-757.e8, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30979586

ABSTRACT

Disturbances in autophagy and stress granule dynamics have been implicated as potential mechanisms underlying inclusion body myopathy (IBM) and related disorders. Yet the roles of core autophagy proteins in IBM and stress granule dynamics remain poorly characterized. Here, we demonstrate that disrupted expression of the core autophagy proteins ULK1 and ULK2 in mice causes a vacuolar myopathy with ubiquitin and TDP-43-positive inclusions; this myopathy is similar to that caused by VCP/p97 mutations, the most common cause of familial IBM. Mechanistically, we show that ULK1/2 localize to stress granules and phosphorylate VCP, thereby increasing VCP's activity and ability to disassemble stress granules. These data suggest that VCP dysregulation and defective stress granule disassembly contribute to IBM-like disease in Ulk1/2-deficient mice. In addition, stress granule disassembly is accelerated by an ULK1/2 agonist, suggesting ULK1/2 as targets for exploiting the higher-order regulation of stress granules for therapeutic intervention of IBM and related disorders.


Subject(s)
Autophagy-Related Protein-1 Homolog/genetics , Lysosomal Storage Diseases/genetics , Muscular Diseases/genetics , Protein Serine-Threonine Kinases/genetics , Valosin Containing Protein/genetics , Adenosine Triphosphatases/genetics , Animals , Autophagy/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Humans , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Mice , Muscular Diseases/metabolism , Muscular Diseases/pathology , Phosphorylation/genetics , Stress, Physiological/genetics , Ubiquitin/genetics
19.
Immunity ; 46(4): 635-648, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28410990

ABSTRACT

Mice carrying a hypomorphic point mutation in the Ptpn6 gene (Ptpn6spin mice) develop an inflammatory skin disease that resembles neutrophilic dermatosis in humans. Here, we demonstrated that interleukin-1α (IL-1α) signaling through IL-1R and MyD88 in both stromal and immune cells drive inflammation in Ptpn6spin mice. We further identified SYK as a critical kinase that phosphorylates MyD88, promoted MyD88-dependent signaling and mediates dermatosis in Ptpn6spin mice. Our studies further demonstrated that SHP1 encoded by Ptpn6 binds and suppresses SYK activation to inhibit MyD88 phosphorylation. Downstream of SHP1 and SYK-dependent counterregulation of MyD88 tyrosine phosphorylation, we have demonstrated that the scaffolding function of receptor interacting protein kinase 1 (RIPK1) and tumor growth factor-ß activated kinase 1 (TAK1)-mediating signaling were required to spur inflammatory disease. Overall, these studies identify SHP1 and SYK crosstalk as a critical regulator of MyD88 post-translational modifications and IL-1-driven inflammation.


Subject(s)
Inflammation/immunology , Interleukin-1alpha/immunology , Myeloid Differentiation Factor 88/immunology , Skin Diseases/immunology , Syk Kinase/immunology , Animals , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Mice, Knockout , Models, Immunological , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Skin Diseases/genetics , Skin Diseases/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism
20.
Nature ; 588(7839): 688-692, 2020 12.
Article in English | MEDLINE | ID: mdl-33268895

ABSTRACT

Inflammasomes are important sentinels of innate immune defence that are activated in response to diverse stimuli, including pathogen-associated molecular patterns (PAMPs)1. Activation of the inflammasome provides host defence against aspergillosis2,3, which is a major health concern for patients who are immunocompromised. However, the Aspergillus fumigatus PAMPs that are responsible for inflammasome activation are not known. Here we show that the polysaccharide galactosaminogalactan (GAG) of A. fumigatus is a PAMP that activates the NLRP3 inflammasome. The binding of GAG to ribosomal proteins inhibited cellular translation machinery, and thus activated the NLRP3 inflammasome. The galactosamine moiety bound to ribosomal proteins and blocked cellular translation, which triggered activation of the NLRP3 inflammasome. In mice, a GAG-deficient Aspergillus mutant (Δgt4c) did not elicit protective activation of the inflammasome, and this strain exhibited enhanced virulence. Moreover, administration of GAG protected mice from colitis induced by dextran sulfate sodium in an inflammasome-dependent manner. Thus, ribosomes connect the sensing of this fungal PAMP to the activation of an innate immune response.


Subject(s)
Aspergillosis/prevention & control , Aspergillus fumigatus/metabolism , Inflammasomes/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Polysaccharides/metabolism , Animals , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillus fumigatus/immunology , Biofilms , Colitis/chemically induced , Colitis/prevention & control , Dextran Sulfate , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Immunity, Innate , Inflammasomes/immunology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polysaccharides/immunology , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL