Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Annu Rev Immunol ; 35: 313-336, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142323

ABSTRACT

Protective immune responses to viral infection are initiated by innate immune sensors that survey extracellular and intracellular space for foreign nucleic acids. The existence of these sensors raises fundamental questions about self/nonself discrimination because of the abundance of self-DNA and self-RNA that occupy these same compartments. Recent advances have revealed that enzymes that metabolize or modify endogenous nucleic acids are essential for preventing inappropriate activation of the innate antiviral response. In this review, we discuss rare human diseases caused by dysregulated nucleic acid sensing, focusing primarily on intracellular sensors of nucleic acids. We summarize lessons learned from these disorders, we rationalize the existence of these diseases in the context of evolution, and we propose that this framework may also apply to a number of more common autoimmune diseases for which the underlying genetics and mechanisms are not yet fully understood.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Autoimmunity , Lupus Erythematosus, Systemic/immunology , Nervous System Malformations/immunology , Nucleic Acids/immunology , Virus Diseases/immunology , Animals , Humans , Immunity, Innate , Interferon Type I/metabolism , Toll-Like Receptors/metabolism
2.
Cell ; 186(25): 5536-5553.e22, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38029747

ABSTRACT

Mycobacterium tuberculosis (Mtb) causes 1.6 million deaths annually. Active tuberculosis correlates with a neutrophil-driven type I interferon (IFN) signature, but the cellular mechanisms underlying tuberculosis pathogenesis remain poorly understood. We found that interstitial macrophages (IMs) and plasmacytoid dendritic cells (pDCs) are dominant producers of type I IFN during Mtb infection in mice and non-human primates, and pDCs localize near human Mtb granulomas. Depletion of pDCs reduces Mtb burdens, implicating pDCs in tuberculosis pathogenesis. During IFN-driven disease, we observe abundant DNA-containing neutrophil extracellular traps (NETs) described to activate pDCs. Cell-type-specific disruption of the type I IFN receptor suggests that IFNs act on IMs to inhibit Mtb control. Single-cell RNA sequencing (scRNA-seq) indicates that type I IFN-responsive cells are defective in their response to IFNγ, a cytokine critical for Mtb control. We propose that pDC-derived type I IFNs act on IMs to permit bacterial replication, driving further neutrophil recruitment and active tuberculosis disease.


Subject(s)
Interferon Type I , Tuberculosis , Humans , Mice , Animals , Macrophages/microbiology , Cytokines , Neutrophils , Dendritic Cells
3.
Immunity ; 57(4): 718-730, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38599167

ABSTRACT

The cGAS-STING intracellular DNA-sensing pathway has emerged as a key element of innate antiviral immunity and a promising therapeutic target. The existence of an innate immune sensor that can be activated by any double-stranded DNA (dsDNA) of any origin raises fundamental questions about how cGAS is regulated and how it responds to "foreign" DNA while maintaining tolerance to ubiquitous self-DNA. In this review, we summarize recent evidence implicating important roles for cGAS in the detection of foreign and self-DNA. We describe two recent and surprising insights into cGAS-STING biology: that cGAS is tightly tethered to the nucleosome and that the cGAMP product of cGAS is an immunotransmitter acting at a distance to control innate immunity. We consider how these advances influence our understanding of the emerging roles of cGAS in the DNA damage response (DDR), senescence, aging, and cancer biology. Finally, we describe emerging approaches to harness cGAS-STING biology for therapeutic benefit.


Subject(s)
Nucleotidyltransferases , Signal Transduction , Nucleotidyltransferases/metabolism , Immunity, Innate , DNA
4.
Immunity ; 55(10): 1799-1812.e4, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36070769

ABSTRACT

The DNA sensor cyclic GMP-AMP synthase (cGAS) is important for antiviral and anti-tumor immunity. cGAS generates cyclic GMP-AMP (cGAMP), a diffusible cyclic dinucleotide that activates the antiviral response through the adaptor protein stimulator of interferon genes (STING). cGAMP cannot passively cross cell membranes, but recent advances have established a role for extracellular cGAMP as an "immunotransmitter" that can be imported into cells. However, the mechanism by which cGAMP exits cells remains unknown. Here, we identifed ABCC1 as a direct, ATP-dependent cGAMP exporter in mouse and human cells. We show that ABCC1 overexpression enhanced cGAMP export and limited STING signaling and that loss of ABCC1 reduced cGAMP export and potentiated STING signaling. We demonstrate that ABCC1 deficiency exacerbated cGAS-dependent autoimmunity in the Trex1-/- mouse model of Aicardi-Goutières syndrome. Thus, ABCC1-mediated cGAMP export is a key regulatory mechanism that limits cell-intrinsic activation of STING and ameliorates STING-dependent autoimmune disease.


Subject(s)
Membrane Transport Proteins , Multidrug Resistance-Associated Proteins , Nucleotides, Cyclic , Adenosine Triphosphate , Animals , DNA/metabolism , Humans , Interferons/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism
5.
Immunity ; 54(9): 1948-1960.e5, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34343497

ABSTRACT

The RNA deaminase ADAR1 is an essential negative regulator of the RNA sensor MDA5, and loss of ADAR1 function triggers inappropriate activation of MDA5 by self-RNAs. Mutations in ADAR, the gene that encodes ADAR1, cause human immune diseases, including Aicardi-Goutières syndrome (AGS). However, the mechanisms of MDA5-dependent disease pathogenesis in vivo remain unknown. Here we generated mice with a single amino acid change in ADAR1 that models the most common human ADAR AGS mutation. These Adar mutant mice developed lethal disease that required MDA5, the RIG-I-like receptor LGP2, type I interferons, and the eIF2α kinase PKR. A small-molecule inhibitor of the integrated stress response (ISR) that acts downstream of eIF2α phosphorylation prevented immunopathology and rescued the mice from mortality. These findings place PKR and the ISR as central components of immunopathology in vivo and identify therapeutic targets for treatment of human diseases associated with the ADAR1-MDA5 axis.


Subject(s)
Adenosine Deaminase/metabolism , Autoimmune Diseases of the Nervous System/pathology , Nervous System Malformations/pathology , Stress, Physiological/physiology , eIF-2 Kinase/metabolism , A549 Cells , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Mutant Strains , Mutation , Nervous System Malformations/genetics , Nervous System Malformations/metabolism
6.
Cell ; 163(7): 1716-29, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26686653

ABSTRACT

Cellular lipid requirements are achieved through a combination of biosynthesis and import programs. Using isotope tracer analysis, we show that type I interferon (IFN) signaling shifts the balance of these programs by decreasing synthesis and increasing import of cholesterol and long chain fatty acids. Genetically enforcing this metabolic shift in macrophages is sufficient to render mice resistant to viral challenge, demonstrating the importance of reprogramming the balance of these two metabolic pathways in vivo. Unexpectedly, mechanistic studies reveal that limiting flux through the cholesterol biosynthetic pathway spontaneously engages a type I IFN response in a STING-dependent manner. The upregulation of type I IFNs was traced to a decrease in the pool size of synthesized cholesterol and could be inhibited by replenishing cells with free cholesterol. Taken together, these studies delineate a metabolic-inflammatory circuit that links perturbations in cholesterol biosynthesis with activation of innate immunity.


Subject(s)
Cholesterol/metabolism , Immunity, Innate , Interferon-gamma/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Humans , Interferon beta-1b , Membrane Proteins/metabolism , Mevalonic Acid/metabolism , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism
7.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553952

ABSTRACT

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Subject(s)
Autoimmunity/genetics , Cardiovirus Infections/genetics , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/genetics , Adolescent , Adult , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/immunology , Blotting, Southern , Cardiovirus Infections/immunology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Encephalomyocarditis virus/immunology , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Immunoblotting , Interferon-Induced Helicase, IFIH1/immunology , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Diseases/genetics , Virus Diseases/immunology , Young Adult
8.
Nature ; 607(7920): 769-775, 2022 07.
Article in English | MEDLINE | ID: mdl-35859177

ABSTRACT

The RNA-editing enzyme ADAR1 is essential for the suppression of innate immune activation and pathology caused by aberrant recognition of self-RNA, a role it carries out by disrupting the duplex structure of endogenous double-stranded RNA species1,2. A point mutation in the sequence encoding the Z-DNA-binding domain (ZBD) of ADAR1 is associated with severe autoinflammatory disease3-5. ZBP1 is the only other ZBD-containing mammalian protein6, and its activation can trigger both cell death and transcriptional responses through the kinases RIPK1 and RIPK3, and the protease caspase 8 (refs. 7-9). Here we show that the pathology caused by alteration of the ZBD of ADAR1 is driven by activation of ZBP1. We found that ablation of ZBP1 fully rescued the overt pathology caused by ADAR1 alteration, without fully reversing the underlying inflammatory program caused by this alteration. Whereas loss of RIPK3 partially phenocopied the protective effects of ZBP1 ablation, combined deletion of caspase 8 and RIPK3, or of caspase 8 and MLKL, unexpectedly exacerbated the pathogenic effects of ADAR1 alteration. These findings indicate that ADAR1 is a negative regulator of sterile ZBP1 activation, and that ZBP1-dependent signalling underlies the autoinflammatory pathology caused by alteration of ADAR1.


Subject(s)
Adenosine Deaminase , Immune System Diseases , Inflammation , Mutation , RNA-Binding Proteins , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Death , Gene Deletion , Immune System Diseases/genetics , Immune System Diseases/metabolism , Immune System Diseases/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mammals/genetics , Protein Kinases/deficiency , Protein Kinases/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
9.
Nat Immunol ; 15(5): 415-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24747712

ABSTRACT

Inappropriate or chronic detection of self nucleic acids by the innate immune system underlies many human autoimmune diseases. We discuss here an unexpected source of endogenous immunostimulatory nucleic acids: the reverse-transcribed cDNA of endogenous retroelements. The interplay between innate immune sensing and clearance of retroelement cDNA has important implications for the understanding of immune responses to infectious retroviruses such as human immunodeficiency virus (HIV). Furthermore, the detection of cDNA by the innate immune system reveals an evolutionary tradeoff: selection for a vigorous, sensitive response to infectious retroviruses may predispose the inappropriate detection of endogenous retroelements. We propose that this tradeoff has placed unique constraints on the sensitivity of the DNA-activated antiviral response, with implications for the interactions of DNA viruses and retroviruses with their hosts. Finally, we discuss how better understanding of the intersection of retroelement biology and innate immunity can guide the way to novel therapies for specific autoimmune diseases.


Subject(s)
Autoimmune Diseases/genetics , DNA, Complementary/immunology , Receptors, Pattern Recognition/immunology , Retroelements/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Biological Evolution , Host-Pathogen Interactions , Humans , Immunity, Innate , Reverse Transcription
10.
Nat Immunol ; 15(9): 839-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25064072

ABSTRACT

Sensors of the innate immune system that detect intracellular nucleic acids must be regulated to prevent inappropriate activation by endogenous DNA and RNA. The exonuclease Trex1 regulates the DNA-sensing pathway by metabolizing potential DNA ligands that trigger it. However, an analogous mechanism for regulating the RIG-I-like receptors (RLRs) that detect RNA remains unknown. We found here that the SKIV2L RNA exosome potently limited the activation of RLRs. The unfolded protein response (UPR), which generated endogenous RLR ligands through the cleavage of cellular RNA by the endonuclease IRE-1, triggered the production of type I interferons in cells depleted of SKIV2L. Humans with deficiency in SKIV2L had a type I interferon signature in their peripheral blood. Our findings reveal a mechanism for the intracellular metabolism of immunostimulatory RNA, with implications for specific autoimmune disorders.


Subject(s)
DEAD-box RNA Helicases/immunology , Diarrhea, Infantile/immunology , Endoribonucleases/immunology , Exosome Multienzyme Ribonuclease Complex , Fetal Growth Retardation/immunology , Hair Diseases/immunology , Immunity, Innate/immunology , Nuclear Proteins/immunology , Protein Serine-Threonine Kinases/immunology , RNA Helicases/immunology , RNA-Binding Proteins/immunology , Unfolded Protein Response/immunology , Animals , DEAD Box Protein 58 , Facies , Gene Knockdown Techniques , Humans , Interferon Type I/immunology , Mice, Inbred C57BL , Proteins/immunology
11.
Immunity ; 45(2): 255-66, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27496731

ABSTRACT

Detection of intracellular DNA triggers activation of the STING-dependent interferon-stimulatory DNA (ISD) pathway, which is essential for antiviral responses. Multiple DNA sensors have been proposed to activate this pathway, including AIM2-like receptors (ALRs). Whether the ALRs are essential for activation of this pathway remains unknown. To rigorously explore the function of ALRs, we generated mice lacking all 13 ALR genes. We found that ALRs are dispensable for the type I interferon (IFN) response to transfected DNA ligands, DNA virus infection, and lentivirus infection. We also found that ALRs do not contribute to autoimmune disease in the Trex1(-/-) mouse model of Aicardi-Goutières Syndrome. Finally, CRISPR-mediated disruption of the human AIM2-like receptor IFI16 in primary fibroblasts revealed that IFI16 is not essential for the IFN response to human cytomegalovirus infection. Our findings indicate that ALRs are dispensable for the ISD response and suggest that alternative functions for these receptors should be explored.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , DNA-Binding Proteins/metabolism , Lentivirus Infections/immunology , Lentivirus/immunology , Nervous System Malformations/immunology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/immunology , DNA-Binding Proteins/genetics , Disease Models, Animal , Exodeoxyribonucleases/genetics , Genetic Loci/genetics , Humans , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Phosphoproteins/genetics
12.
Immunity ; 43(5): 933-44, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588779

ABSTRACT

Mutations in ADAR, which encodes the ADAR1 RNA-editing enzyme, cause Aicardi-Goutières syndrome (AGS), a severe autoimmune disease associated with an aberrant type I interferon response. How ADAR1 prevents autoimmunity remains incompletely defined. Here, we demonstrate that ADAR1 is a specific and essential negative regulator of the MDA5-MAVS RNA sensing pathway. Moreover, we uncovered a MDA5-MAVS-independent function for ADAR1 in the development of multiple organs. We showed that the p150 isoform of ADAR1 uniquely regulated the MDA5 pathway, whereas both the p150 and p110 isoforms contributed to development. Abrupt deletion of ADAR1 in adult mice revealed that both of these functions were required throughout life. Our findings delineate genetically separable roles for both ADAR1 isoforms in vivo, with implications for the human diseases caused by ADAR mutations.


Subject(s)
Adenosine Deaminase/metabolism , Autoimmunity/physiology , DEAD-box RNA Helicases/metabolism , Protein Isoforms/metabolism , RNA Editing/physiology , RNA/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmune Diseases of the Nervous System/metabolism , HEK293 Cells , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1 , Mice , Nervous System Malformations/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/physiology
13.
Cell ; 134(4): 587-98, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18724932

ABSTRACT

Detection of nucleic acids and induction of type I interferons (IFNs) are principal elements of antiviral defense but can cause autoimmunity if misregulated. Cytosolic DNA detection activates a potent, cell-intrinsic antiviral response through a poorly defined pathway. In a screen for proteins relevant to this IFN-stimulatory DNA (ISD) response, we identify 3' repair exonuclease 1 (Trex1). Mutations in the human trex1 gene cause Aicardi-Goutieres syndrome (AGS) and chilblain lupus, but the molecular basis of these diseases is unknown. We define Trex1 as an essential negative regulator of the ISD response and delineate the genetic pathway linking Trex1 deficiency to lethal autoimmunity. We show that single-stranded DNA derived from endogenous retroelements accumulates in Trex1-deficient cells, and that Trex1 can metabolize reverse-transcribed DNA. These findings reveal a cell-intrinsic mechanism for initiation of autoimmunity, implicate the ISD pathway as the cause of AGS, and suggest an unanticipated contribution of endogenous retroelements to autoimmunity.


Subject(s)
Autoimmunity , DNA, Single-Stranded/immunology , Exodeoxyribonucleases/immunology , Phosphoproteins/immunology , Retroelements/genetics , Animals , Autoantibodies/immunology , Cell Line , Exodeoxyribonucleases/genetics , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Mice , Phosphoproteins/genetics , Retroelements/immunology
14.
Immunity ; 36(1): 120-31, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22284419

ABSTRACT

The type I interferon (IFN) response initiated by detection of nucleic acids is important for antiviral defense but is also associated with specific autoimmune diseases. Mutations in the human 3' repair exonuclease 1 (Trex1) gene cause Aicardi-Goutières syndrome (AGS), an IFN-associated autoimmune disease. However, the source of the type I IFN response and the precise mechanisms of disease in AGS remain unknown. Here, we demonstrate that Trex1 is an essential negative regulator of the STING-dependent antiviral response. We used an in vivo reporter of IFN activity in Trex1-deficient mice to localize the initiation of disease to nonhematopoietic cells. These IFNs drove T cell-mediated inflammation and an autoantibody response that targeted abundant, tissue-restricted autoantigens. However, B cells contributed to mortality independently of T cell-mediated tissue damage. These findings reveal a stepwise progression of autoimmune disease in Trex1-deficient mice, with implications for the treatment of AGS and related disorders.


Subject(s)
Autoimmune Diseases/physiopathology , Autoimmunity/immunology , Interferons , Lymphocytes/immunology , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases of the Nervous System/physiopathology , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Interferons/physiology , Mice , Mice, Knockout , Models, Biological , Nervous System Malformations/physiopathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 115(26): 6798-6803, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891701

ABSTRACT

Detection of nucleic acids by innate immune sensors triggers the production of type I interferons (IFNs). While IFNs are essential for host defense against viral infection, dysregulated production of IFNs underlies numerous autoinflammatory diseases. We have found that the loss of sumoylation results in a potent, spontaneous IFN response. Vertebrates possess three small ubiquitin-like modifiers (SUMOs) that can be conjugated onto target proteins and alter protein function in diverse but still poorly characterized ways. We demonstrate that regulation of IFN by sumoylation is redundantly mediated by both SUMO2 and SUMO3, but not SUMO1, revealing a previously unknown function of SUMO2/3. Remarkably, this IFN response is independent of all known IFN-inducing pathways and does not require either of the canonical IFN-associated transcription factors IRF3 or IRF7. Taken together, our findings demonstrate that SUMO2 and SUMO3 are specific and essential negative regulators of a noncanonical mechanism of IFN induction.


Subject(s)
Interferon Type I/metabolism , Signal Transduction/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation/physiology , Ubiquitins/metabolism , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Type I/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , THP-1 Cells , Ubiquitins/genetics
16.
J Immunol ; 200(8): 2748-2756, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29540580

ABSTRACT

The sensing of viral nucleic acids within the cytosol is essential for the induction of innate immune responses following infection. However, this sensing occurs within cells that have already been infected. The death of infected cells can be beneficial to the host by eliminating the virus's replicative niche and facilitating the release of inflammatory mediators. In this study, we show that sensing of intracellular DNA or RNA by cGAS-STING or RIG-I-MAVS, respectively, leads to activation of RIPK3 and necroptosis in bone marrow-derived macrophages. Notably, this requires signaling through both type I IFN and TNF receptors, revealing synergy between these pathways to induce cell death. Furthermore, we show that hyperactivation of STING in mice leads to a shock-like phenotype, the mortality of which requires activation of the necroptotic pathway and IFN and TNF cosignaling, demonstrating that necroptosis is one outcome of STING signaling in vivo.


Subject(s)
Cell Death/physiology , Interferon Type I/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , DNA, Viral/immunology , Immunity, Innate/immunology , Interferon Type I/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , RNA, Viral/immunology , Tumor Necrosis Factor-alpha/immunology , Virus Diseases/immunology
17.
J Immunol ; 195(5): 1939-43, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26223655

ABSTRACT

Detection of intracellular DNA triggers activation of the stimulator of IFN genes-dependent IFN-stimulatory DNA (ISD) pathway, which is essential for antiviral immune responses. However, chronic activation of this pathway is implicated in autoimmunity. Mutations in TREX1, a 3' repair exonuclease that degrades cytosolic DNA, cause Aicardi-Goutières syndrome and chilblain lupus. Trex1 (-/-) mice develop lethal, IFN-driven autoimmune disease that is dependent on activation of the ISD pathway, but the DNA sensors that detect the endogenous DNA that accumulates in Trex1 (-/-) mice have not been defined. Multiple DNA sensors have been proposed to activate the ISD pathway, including cyclic GMP-AMP synthase (cGAS). In this study, we show that Trex1 (-/-) mice lacking cGAS are completely protected from lethality, exhibit dramatically reduced tissue inflammation, and fail to develop autoantibodies. These findings implicate cGAS as a key driver of autoimmune disease and suggest that cGAS inhibitors may be useful therapeutics for Aicardi-Goutières syndrome and related autoimmune diseases.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Exodeoxyribonucleases/immunology , Nervous System Malformations/immunology , Nucleotidyltransferases/immunology , Phosphoproteins/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian/cytology , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression/drug effects , Gene Expression/immunology , Humans , Immunoblotting , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Interferons/immunology , Interferons/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nucleotides, Cyclic/immunology , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Nat Struct Mol Biol ; 30(1): 72-80, 2023 01.
Article in English | MEDLINE | ID: mdl-36593311

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is a pattern recognition receptor critical for the innate immune response to intracellular pathogens, DNA damage, tumorigenesis and senescence. Binding to double-stranded DNA (dsDNA) induces conformational changes in cGAS that activate the enzyme to produce 2'-3' cyclic GMP-AMP (cGAMP), a second messenger that initiates a potent interferon (IFN) response through its receptor, STING. Here, we combined two-state computational design with informatics-guided design to create constitutively active, dsDNA ligand-independent cGAS (CA-cGAS). We identified CA-cGAS mutants with IFN-stimulating activity approaching that of dsDNA-stimulated wild-type cGAS. DNA-independent adoption of the active conformation was directly confirmed by X-ray crystallography. In vivo expression of CA-cGAS in tumor cells resulted in STING-dependent tumor regression, demonstrating that the designed proteins have therapeutically relevant biological activity. Our work provides a general framework for stabilizing active conformations of enzymes and provides CA-cGAS variants that could be useful as genetically encoded adjuvants and tools for understanding inflammatory diseases.


Subject(s)
Immunity, Innate , Nucleotidyltransferases , Nucleotidyltransferases/metabolism , DNA/chemistry
19.
J Exp Med ; 203(8): 1837-41, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16880263

ABSTRACT

Mice lacking the adaptor protein that initiates an antiviral response downstream of the RNA helicases retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) have recently been described. These studies highlight the essential and nonredundant role of nucleic acid recognition in the induction of type I interferon production and raise important questions regarding the nature of cell-autonomous virus detection in coordinating the antiviral response.


Subject(s)
Antiviral Agents/metabolism , Interferon Type I/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Mice , Nucleic Acids/metabolism
20.
Nat Rev Immunol ; 22(8): 471-483, 2022 08.
Article in English | MEDLINE | ID: mdl-34671122

ABSTRACT

As brutally demonstrated by the COVID-19 pandemic, an effective immune system is essential for survival. Developed over evolutionary time, viral nucleic acid detection is a central pillar in the defensive armamentarium used to combat foreign microbial invasion. To ensure cellular homeostasis, such a strategy necessitates the efficient discrimination of pathogen-derived DNA and RNA from that of the host. In 2011, it was suggested that an upregulation of type I interferon signalling might serve as a defining feature of a novel set of Mendelian inborn errors of immunity, where antiviral sensors are triggered by host nucleic acids due to a failure of self versus non-self discrimination. These rare disorders have played a surprisingly significant role in informing our understanding of innate immunity and the relevance of type I interferon signalling for human health and disease. Here we consider what we have learned in this time, and how the field may develop in the future.


Subject(s)
COVID-19 , Interferon Type I , Nucleic Acids , Humans , Immunity, Innate , Interferon Type I/genetics , Pandemics
SELECTION OF CITATIONS
SEARCH DETAIL