Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Front Immunol ; 13: 826880, 2022.
Article in English | MEDLINE | ID: mdl-35185917

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) recognizes double-stranded DNA (dsDNA) derived from invading pathogens and induces an interferon response via activation of the key downstream adaptor protein stimulator of interferon genes (STING). This is the most classic biological function of the cGAS-STING signaling pathway and is critical for preventing pathogenic microorganism invasion. In addition, cGAS can interact with various types of nucleic acids, including cDNA, DNA : RNA hybrids, and circular RNA, to contribute to a diverse set of biological functions. An increasing number of studies have revealed an important relationship between the cGAS-STING signaling pathway and autophagy, cellular senescence, antitumor immunity, inflammation, and autoimmune diseases. This review details the mechanism of action of cGAS as it interacts with different types of nucleic acids, its rich biological functions, and the potential for targeting this pathway to treat various diseases.


Subject(s)
Inflammation/etiology , Membrane Proteins/physiology , Nucleic Acids/classification , Nucleotidyltransferases/physiology , Animals , Autoimmune Diseases/immunology , Autophagy/physiology , Cellular Senescence , DNA/metabolism , Humans , Interferon Type I/physiology , Signal Transduction/physiology
2.
Life Sci ; 291: 120263, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34971697

ABSTRACT

AIMS: Myocardial infarction (MI) is a major global cause of death. Massive cell death leads to inflammation, which is necessary for ensuing wound healing. Extensive inflammation, however, promotes infarct expansion and adverse remodeling. The DNA sensing receptor cyclic GMP-AMP synthase and its downstream signaling effector stimulator of interferon genes (cGAS-STING) is central in innate immune reactions in infections or autoimmunity. Cytosolic double-strand DNA activates the pathway and down-stream inflammatory responses. Recent papers demonstrated that this pathway is also active following MI and that its genetic targeting improves outcome. Thus, we investigated if pharmacologic pathway inhibition is protective after MI in order to test its translational potential. MAIN METHODS: We investigated novel and selective small-molecule STING inhibitors that inhibit STING palmitoylation and multimerization and thereby downstream pathway activation in a preclinical murine MI model. We assessed structural and functional cardiac remodeling, infarct expansion and fibrosis, as well as cardiomyocyte hypertrophy and the expression of inflammatory genes. KEY FINDINGS: Pharmacologic STING inhibition did not reduce mortality due to myocardial rupture in non-reperfused MI. Infarct size at day one was comparable. However, three weeks of pharmacologic STING inhibition after reperfused MI decreased infarct expansion and scarring, increased left ventricular systolic function to levels approaching normal values, and reduced myocardial hypertrophy. SIGNIFICANCE: Selective small-molecule STING inhibition after myocardial infarction has the potential to improve wound healing responses and pathological remodeling and thereby attenuate the development of ischemic heart failure.


Subject(s)
Membrane Proteins/metabolism , Myocardial Infarction/metabolism , Nucleotidyltransferases/metabolism , Animals , Heart/physiopathology , Heart Failure/physiopathology , Inflammation/pathology , Lipoylation/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Nucleotidyltransferases/physiology , Signal Transduction , Systole , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology
3.
Cell Rep ; 37(6): 109977, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758308

ABSTRACT

Tumor necrosis factor (TNF) is a key driver of several inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis, in which affected tissues show an interferon-stimulated gene signature. Here, we demonstrate that TNF triggers a type-I interferon response that is dependent on the cyclic guanosine monophosphate-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. We show that TNF inhibits PINK1-mediated mitophagy and leads to altered mitochondrial function and to an increase in cytosolic mtDNA levels. Using cGAS-chromatin immunoprecipitation (ChIP), we demonstrate that cytosolic mtDNA binds to cGAS after TNF treatment. Furthermore, TNF induces a cGAS-STING-dependent transcriptional response that mimics that of macrophages from rheumatoid arthritis patients. Finally, in an inflammatory arthritis mouse model, cGAS deficiency blocked interferon responses and reduced inflammatory cell infiltration and joint swelling. These findings elucidate a molecular mechanism linking TNF to type-I interferon signaling and suggest a potential benefit for therapeutic targeting of cGAS/STING in TNF-driven diseases.


Subject(s)
Arthritis, Experimental/immunology , DNA, Mitochondrial/metabolism , Immunity, Innate , Inflammation/immunology , Interferon Type I/pharmacology , Membrane Proteins/metabolism , Nucleotidyltransferases/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , DNA, Mitochondrial/drug effects , Female , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitophagy
4.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34819364

ABSTRACT

Mitotic errors can activate cyclic GMP-AMP synthase (cGAS) and induce type I interferon (IFN) signaling. Current models propose that chromosome segregation errors generate micronuclei whose rupture activates cGAS. We used a panel of antimitotic drugs to perturb mitosis in human fibroblasts and measured abnormal nuclear morphologies, cGAS localization, and IFN signaling in the subsequent interphase. Micronuclei consistently recruited cGAS without activating it. Instead, IFN signaling correlated with formation of cGAS-coated chromatin bridges that were selectively generated by microtubule stabilizers and MPS1 inhibitors. cGAS activation by chromatin bridges was suppressed by drugs that prevented cytokinesis. We confirmed cGAS activation by chromatin bridges in cancer lines that are unable to secrete IFN by measuring paracrine transfer of 2'3'-cGAMP to fibroblasts, and in mouse cells. We propose that cGAS is selectively activated by self-chromatin when it is stretched in chromatin bridges. Immunosurveillance of cells that fail mitosis, and antitumor actions of taxanes and MPS1 inhibitors, may depend on this effect.


Subject(s)
Chromatin/physiology , Mitosis/physiology , Nucleotidyltransferases/metabolism , Cell Line, Tumor , Chromatin/genetics , Humans , Interferon Type I/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Micronucleus, Germline/genetics , Micronucleus, Germline/physiology , Mitosis/drug effects , Mitosis/genetics , Neoplasms/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/physiology , Signal Transduction
5.
Front Immunol ; 12: 753789, 2021.
Article in English | MEDLINE | ID: mdl-34659260

ABSTRACT

Innate immunity is regulated by a broad set of evolutionary conserved receptors to finely probe the local environment and maintain host integrity. Besides pathogen recognition through conserved motifs, several of these receptors also sense aberrant or misplaced self-molecules as a sign of perturbed homeostasis. Among them, self-nucleic acid sensing by the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway alerts on the presence of both exogenous and endogenous DNA in the cytoplasm. We review recent literature demonstrating that self-nucleic acid detection through the STING pathway is central to numerous processes, from cell physiology to sterile injury, auto-immunity and cancer. We address the role of STING in autoimmune diseases linked to dysfunctional DNAse or related to mutations in DNA sensing pathways. We expose the role of the cGAS/STING pathway in inflammatory diseases, neurodegenerative conditions and cancer. Connections between STING in various cell processes including autophagy and cell death are developed. Finally, we review proposed mechanisms to explain the sources of cytoplasmic DNA.


Subject(s)
Autoimmune Diseases/immunology , DNA/analysis , Immunity, Innate/physiology , Inflammation/immunology , Membrane Proteins/physiology , Neoplasms/immunology , Neurodegenerative Diseases/immunology , Adenosine Triphosphate/metabolism , Adult , Autoimmune Diseases/physiopathology , Autophagy , Cytokines/physiology , Cytoplasm/chemistry , Guanosine Triphosphate/metabolism , Humans , Infant , Inflammation/physiopathology , Interferon Type I/physiology , Mitochondria/physiology , NF-kappa B/metabolism , Neoplasms/physiopathology , Neurodegenerative Diseases/physiopathology , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/physiology , Signal Transduction/physiology
6.
Commun Biol ; 4(1): 820, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188173

ABSTRACT

Host lipid metabolism and viral responses are intimately connected. However, the process by which the acquired immune systems adapts lipid metabolism to meet demands, and whether or not the metabolic rewiring confers a selective advantage to host immunity, remains unclear. Here we show that viral infection attenuates the expression of genes related to lipid metabolism in murine CD4+ T cells, which in turn increases the expression of antiviral genes. Inhibition of the fatty acid synthesis pathway substantially increases the basal expression of antiviral genes via the spontaneous production of type I interferon (IFN). Using a combination of CRISPR/Cas9-mediated genome editing technology and a global lipidomics analysis, we found that the decrease in monounsaturated fatty acid caused by genetic deletion of Scd2 in mice was crucial for the induction of an antiviral response through activation of the cGAS-STING pathway. These findings demonstrate the important relationship between fatty acid biosynthesis and type I IFN responses that enhances the antiviral response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fatty Acids, Monounsaturated/metabolism , Interferon Type I/pharmacology , Membrane Proteins/physiology , Nucleotidyltransferases/physiology , Stearoyl-CoA Desaturase/physiology , Virus Diseases/immunology , Animals , Host-Pathogen Interactions , Lipid Metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , Virus Diseases/metabolism
7.
Cell Immunol ; 362: 104298, 2021 04.
Article in English | MEDLINE | ID: mdl-33592541

ABSTRACT

Myeloid derived suppressor cells (MDSCs) are a highly heterogeneous population of immature immune cells with immunosuppressive functions that are recruited to the tumor microenvironment (TME). MDSCs promote tumor growth and progression by inhibiting immune effector cell proliferation and function. MDSCs are affected by both novel anti-cancer therapies targeting the immune system to promote anti-tumor immunity, as well as by conventional treatments such as radiotherapy. Following radiotherapy, cytoplasmic double stranded DNA stimulates the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway, resulting in type I interferon production. Effectiveness of radiotherapy and cGAS/STING signaling are closely intertwined: activation of cGAS and STING is key to generate systemic anti-tumor immunity after irradiation. This review focuses on how radiotherapy and cGAS/STING signaling in MDSCs and/or tumor cells impact MDSC recruitment, expansion and function. The influence of conventional and ablative radiotherapy treatment schedules, inflammatory response following radiotherapy, and hypoxia are discussed as MDSC modulators.


Subject(s)
Membrane Proteins/metabolism , Myeloid-Derived Suppressor Cells/immunology , Nucleotidyltransferases/metabolism , Humans , Immunity, Innate , Interferon Type I/immunology , Interferon Type I/metabolism , Membrane Proteins/physiology , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/pathology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/physiology , Radiotherapy/methods , Signal Transduction/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology
8.
Cell Rep ; 33(10): 108490, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33296647

ABSTRACT

Cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS), upon sensing cytosolic DNA, catalyzes the production of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which activates STING-TBK1-IRF3 signaling. cGAS is also present in the nucleus, but the relevant nuclear function or mechanism remains largely unknown. Here, we report that nuclear cGAS is indispensable for inducing cytokines and chemokines triggered by RNA/DNA viruses. Unexpectedly, the DNA-binding/nucleotidyltransferase activity of cGAS is dispensable for RNA-virus-induced genes expression. cGAS deficiency does not affect the phosphorylation, dimerization, or nuclear translocation of IRF3 induced by double-stranded RNA (dsRNA). Mechanistically, nuclear-localized cGAS interacts with protein arginine methyltransferase 5 (Prmt5), which catalyzes the symmetric dimethylation of histone H3 arginine 2 at Ifnb and Ifna4 promoters, thus facilitating the access of IRF3. Deficiency of Prmt5 or disrupting its catalytic activity suppresses the production of type I interferons (IFNs), impairing the host defenses against RNA/DNA virus infections. Taken together, our study uncovers a non-canonical function of nuclear-localized cGAS in innate immunity via regulating histone arginine modification.


Subject(s)
Nucleotidyltransferases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Antiviral Agents/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic GMP/metabolism , Cytosol/metabolism , Female , Humans , Immunity, Innate/genetics , Interferon Type I/metabolism , Interferon-beta/metabolism , Male , Membrane Proteins/metabolism , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotides, Cyclic , Nucleotidyltransferases/physiology , Phosphorylation/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/physiology , Signal Transduction/physiology
9.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 36(4): 336-339, 2020 Jul.
Article in Chinese | MEDLINE | ID: mdl-33167094

ABSTRACT

OBJECTIVE: To investigate the effects of cyclic GMP-AMP synthase (cGAS) on the proliferation, migration and epithelial to mesenchymal transition (EMT) of breast cancer cells. METHODS: The cGAS lentiviral vector and control fluorescence vector were transfected into breast cancer MCF7 cells and were divided into negative group (NC) and MCF7-cGAS group. The effect of cGAS on proliferation in the MCF7 cells was detected by MTT. The effect of cGAS on cell migration was detected by Transwell assay. The expressions of EMT related proteins were analyzed by Western blot. RESULTS: After over-expressed with cGAS, the proliferation and migration of MCF7 cells were increased (P<0.05). The expression level of the epithelial markers E-cadherin was decreased, while the expression level of the mesenchymal markers N-cadherin was increased(P<0.05). CONCLUSION: The over-expression of cGAS increased the proliferation and migration of breast cancer cells and induced EMT in breast cancer cells.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Nucleotidyltransferases , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Nucleotides, Cyclic , Nucleotidyltransferases/physiology
10.
Cell Rep ; 33(3): 108297, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086059

ABSTRACT

The immune system is not only required for preventing threats exerted by pathogens but also essential for developing immune tolerance to avoid tissue damage. This study identifies a distinct mechanism by which MYSM1 suppresses innate immunity and autoimmunity. The expression of MYSM1 is induced upon DNA virus infection and by intracellular DNA stimulation. MYSM1 subsequently interacts with STING and cleaves STING K63-linked ubiquitination to suppress cGAS-STING signaling. Notably, Mysm1-deficient mice exhibit a hyper-inflammatory response, acute tissue damage, and high mortality upon virus infection. Moreover, in the PBMCs of patients with systemic lupus erythematosus (SLE), MYSM1 production decreases, while type I interferons and pro-inflammatory cytokine expressions increase. Importantly, MYSM1 treatment represses the production of IFNs and pro-inflammatory cytokines in the PBMCs of SLE patients. Thus, MYSM1 is a critical repressor of innate immunity and autoimmunity and is thus a potential therapeutic agent for infectious, inflammatory, and autoimmune diseases.


Subject(s)
Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/metabolism , Adult , Animals , Autoimmune Diseases , Autoimmunity/immunology , China , Female , Humans , Immunity, Innate/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon Type I/physiology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nucleotidyltransferases/physiology , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology
11.
Mol Cell ; 78(6): 1237-1251.e7, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32442397

ABSTRACT

DNA replication stress can stall replication forks, leading to genome instability. DNA damage tolerance pathways assist fork progression, promoting replication fork reversal, translesion DNA synthesis (TLS), and repriming. In the absence of the fork remodeler HLTF, forks fail to slow following replication stress, but underlying mechanisms and cellular consequences remain elusive. Here, we demonstrate that HLTF-deficient cells fail to undergo fork reversal in vivo and rely on the primase-polymerase PRIMPOL for repriming, unrestrained replication, and S phase progression upon limiting nucleotide levels. By contrast, in an HLTF-HIRAN mutant, unrestrained replication relies on the TLS protein REV1. Importantly, HLTF-deficient cells also exhibit reduced double-strand break (DSB) formation and increased survival upon replication stress. Our findings suggest that HLTF promotes fork remodeling, preventing other mechanisms of replication stress tolerance in cancer cells. This remarkable plasticity of the replication fork may determine the outcome of replication stress in terms of genome integrity, tumorigenesis, and response to chemotherapy.


Subject(s)
DNA Replication/physiology , DNA-Binding Proteins/metabolism , DNA/biosynthesis , Transcription Factors/metabolism , Cell Line, Tumor , DNA/genetics , DNA Damage/genetics , DNA Primase/metabolism , DNA Primase/physiology , DNA Repair/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/physiology , HEK293 Cells , Humans , K562 Cells , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/physiology , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/physiology , Transcription Factors/genetics
12.
Front Immunol ; 11: 615, 2020.
Article in English | MEDLINE | ID: mdl-32411126

ABSTRACT

Double-stranded DNA (dsDNA) sensor cyclic-GMP-AMP synthase (cGAS) along with the downstream stimulator of interferon genes (STING) acting as essential immune-surveillance mediators have become hot topics of research. The intrinsic function of the cGAS-STING pathway facilitates type-I interferon (IFN) inflammatory signaling responses and other cellular processes such as autophagy, cell survival, senescence. cGAS-STING pathway interplays with other innate immune pathways, by which it participates in regulating infection, inflammatory disease, and cancer. The therapeutic approaches targeting this pathway show promise for future translation into clinical applications. Here, we present a review of the important previous works and recent advances regarding the cGAS-STING pathway, and provide a comprehensive understanding of the modulatory pattern of the cGAS-STING pathway under multifarious pathologic states.


Subject(s)
Inflammation/etiology , Membrane Proteins/physiology , Nucleotidyltransferases/physiology , Autophagy/physiology , Cell Survival , Cellular Senescence , DNA/metabolism , Humans , Infections/immunology , Interferon Type I/physiology , Neoplasms/immunology , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Aging (Albany NY) ; 12(8): 6904-6927, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32310823

ABSTRACT

Gastric cancer (GC) is one of the most commonly diagnosed malignancies in digestive tract and its underlying molecular mechanism is still not clear, so we aimed to reveal the relationship between GC and UDP-GlcNAc pyrophosphorylase-1 like 1 (UAP1L1). The detection of UAP1L1 expression in GC tumor and normal tissues was accomplished by immunohistochemistry and demonstrated the upregulation of UAP1L1 in GC, which was statistically associated with tumor grade. GC cell models constructed via transfection of UAP1L1-silencing/overexpressing lentiviruses were employed for evaluating the effects of UAP1L1 knockdown/overexpression on GC in vitro and in vivo. The results indicated that UAP1L1 played important role in development of GC through regulating cell proliferation, colony formation, cell apoptosis and cell migration. Subsequently, CDK6 was identified as a potential target in UAP1L1 induced regulation of GC, downregulation of which exhibited similar inhibition effects on GC with UAP1L1. Moreover, it was demonstrated that the promotion of GC by UAP1L1 overexpression could be significantly attenuated or even reversed by simultaneously silencing CDK6. In conclusion, UAP1L1 was reported to be a tumor promotor in the development and progression of GC which may exert its role through regulating CDK6 and may act as a candidate of therapeutic target in treatment.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Nucleotidyltransferases/physiology , Stomach Neoplasms/etiology , Adult , Aged , Animals , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Middle Aged , Nucleotidyltransferases/analysis , Nucleotidyltransferases/genetics , Stomach Neoplasms/chemistry , Up-Regulation
14.
PLoS Pathog ; 15(10): e1007778, 2019 10.
Article in English | MEDLINE | ID: mdl-31603920

ABSTRACT

Type I interferons (T1-IFN) are critical in the innate immune response, acting upon infected and uninfected cells to initiate an antiviral state by expressing genes that inhibit multiple stages of the lifecycle of many viruses. T1-IFN triggers the production of Interferon-Stimulated Genes (ISGs), activating an antiviral program that reduces virus replication. The importance of the T1-IFN response is highlighted by the evolution of viral evasion strategies to inhibit the production or action of T1-IFN in virus-infected cells. T1-IFN is produced via activation of pathogen sensors within infected cells, a process that is targeted by virus-encoded immunomodulatory molecules. This is probably best exemplified by the prototypic poxvirus, Vaccinia virus (VACV), which uses at least 6 different mechanisms to completely block the production of T1-IFN within infected cells in vitro. Yet, mice lacking aspects of T1-IFN signaling are often more susceptible to infection with many viruses, including VACV, than wild-type mice. How can these opposing findings be rationalized? The cytosolic DNA sensor cGAS has been implicated in immunity to VACV, but has yet to be linked to the production of T1-IFN in response to VACV infection. Indeed, there are two VACV-encoded proteins that effectively prevent cGAS-mediated activation of T1-IFN. We find that the majority of VACV-infected cells in vivo do not produce T1-IFN, but that a small subset of VACV-infected cells in vivo utilize cGAS to sense VACV and produce T1-IFN to protect infected mice. The protective effect of T1-IFN is not mediated via ISG-mediated control of virus replication. Rather, T1-IFN drives increased expression of CCL4, which recruits inflammatory monocytes that constrain the VACV lesion in a virus replication-independent manner by limiting spread within the tissue. Our findings have broad implications in our understanding of pathogen detection and viral evasion in vivo, and highlight a novel immune strategy to protect infected tissue.


Subject(s)
Chemokine CCL4/metabolism , Interferon Type I/pharmacology , Membrane Proteins/physiology , Nucleotidyltransferases/physiology , Vaccinia virus/drug effects , Vaccinia/prevention & control , Viral Load/drug effects , Animals , Antiviral Agents/pharmacology , Chemokine CCL4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/virology , Vaccinia/immunology , Vaccinia/metabolism , Vaccinia/virology , Vaccinia virus/immunology , Virus Replication
15.
EMBO J ; 38(21): e102718, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31544964

ABSTRACT

DNA repair via homologous recombination (HR) is indispensable for genome integrity and cell survival but if unrestrained can result in undesired chromosomal rearrangements. The regulatory mechanisms of HR are not fully understood. Cyclic GMP-AMP synthase (cGAS) is best known as a cytosolic innate immune sensor critical for the outcome of infections, inflammatory diseases, and cancer. Here, we report that cGAS is primarily a chromatin-bound protein that inhibits DNA repair by HR, thereby accelerating genome destabilization, micronucleus generation, and cell death under conditions of genomic stress. This function is independent of the canonical STING-dependent innate immune activation and is physiologically relevant for irradiation-induced depletion of bone marrow cells in mice. Mechanistically, we demonstrate that inhibition of HR repair by cGAS is linked to its ability to self-oligomerize, causing compaction of bound template dsDNA into a higher-ordered state less amenable to strand invasion by RAD51-coated ssDNA filaments. This previously unknown role of cGAS has implications for understanding its involvement in genome instability-associated disorders including cancer.


Subject(s)
Cell Death , Cell Nucleus/metabolism , Chromatin/metabolism , Genomic Instability , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/physiology , Recombinational DNA Repair , Animals , Cell Nucleus/genetics , Chromatin/genetics , DNA Damage , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/genetics , Signal Transduction
16.
Radiother Oncol ; 141: 262-266, 2019 12.
Article in English | MEDLINE | ID: mdl-31439450

ABSTRACT

The cGAS-STING pathway can be activated by radiation induced DNA damage and because of its important role in anti-cancer immunity activation, methods to increase its activation in cancer cells could provide significant therapeutic benefits for patients. We explored the impact of hafnium oxide nanoparticles (NBTXR3) activated by radiotherapy on cell death, DNA damage, and activation of the cGAS-STING pathway. We demonstrate that NBTXR3 activated by radiotherapy enhances cell destruction, DNA double strand breaks, micronuclei formation and cGAS-STING pathway activation in a human colorectal cancer model, compared to radiotherapy alone.


Subject(s)
Colorectal Neoplasms/radiotherapy , DNA Damage , Hafnium/pharmacology , Membrane Proteins/physiology , Nanoparticles , Nucleotidyltransferases/physiology , Oxides/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Signal Transduction/drug effects
17.
Cell Rep ; 27(9): 2649-2664.e5, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31141689

ABSTRACT

Lung inflammation induced by silica impairs host control of tuberculosis, yet the underlying mechanism remains unclear. Here, we show that silica-driven exacerbation of M. tuberculosis infection associates with raised type 2 immunity. Silica increases pulmonary Th2 cell and M2 macrophage responses, while reducing type 1 immunity after M. tuberculosis infection. Silica induces lung damage that prompts extracellular self-DNA release and activates STING. This STING priming potentiates M. tuberculosis DNA sensing by and activation of cGAS/STING, which triggers enhanced type I interferon (IFNI) response and type 2 immunity. cGAS-, STING-, and IFNAR-deficient mice are resistant to silica-induced exacerbation of M. tuberculosis infection. Thus, silica-induced self-DNA primes the host response to M. tuberculosis-derived nucleic acids, which increases type 2 immunity while reducing type 1 immunity, crucial for controlling M. tuberculosis infection. These data show how cGAS/STING pathway activation, at the crossroads of sterile inflammation and infection, may affect the host response to pathogens such as M. tuberculosis.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Membrane Proteins/physiology , Mycobacterium tuberculosis/immunology , Pneumonia/complications , Silicon Dioxide/toxicity , Tuberculosis/etiology , Animals , Dendritic Cells , Interferon Regulatory Factor-3/physiology , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/physiology , Pneumonia/chemically induced , Receptor, Interferon alpha-beta/physiology , Signal Transduction , Tuberculosis/metabolism , Tuberculosis/pathology
18.
FEBS J ; 286(14): 2711-2725, 2019 07.
Article in English | MEDLINE | ID: mdl-30963698

ABSTRACT

Rev1, a Y-family DNA polymerase, is involved in the tolerance of DNA damage by translesion DNA synthesis (TLS). Previous studies have shown that the C-terminal domain (CTD) and ubiquitin (Ub)-binding (UBM) domains of Rev1 play important roles in UV-damage tolerance, but how these domains contribute to the process remains unclear. In this study, we created Ub mutations in a proliferating cell nuclear antigen (PCNA)-Ub fusion that differentially affect its interaction with Rev1 and Polη and found that UV-damage tolerance depends on its interaction with Rev1 but not Polη. We also created Rev1-UBM mutations altering its interaction with a PCNA-Ub fusion and Rev1-CTD mutations affecting its interaction with Polη and the Rev7 subunit of Polζ. We thus demonstrated that elevated expression of Rev1 alone is sufficient to confer enhanced UV-damage tolerance and that this tolerance depends on its physical interaction with monoubiquitinated PCNA and Polζ but is independent of Polη. Collectively, these studies reveal central roles played by Rev1 in coordinating UV-damage response pathway choice in mammalian cells.


Subject(s)
DNA Damage , Nucleotidyltransferases/physiology , Ultraviolet Rays , DNA-Directed DNA Polymerase/physiology , HCT116 Cells , Humans , Mutation , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Proliferating Cell Nuclear Antigen/physiology , Ubiquitin/physiology
19.
J Leukoc Biol ; 106(1): 171-185, 2019 07.
Article in English | MEDLINE | ID: mdl-30990921

ABSTRACT

Various intracellular pattern recognition receptors (PRRs) recognize cytosolic pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Cyclic GMP-AMP synthase (cGAS), a cytosolic PRR, recognizes cytosolic nucleic acids including dsDNAs. The recognition of dsDNA by cGAS generates cyclic GMP-AMP (GAMP). The cGAMP is then recognized by STING generating type 1 IFNs and NF-κB-mediated generation of pro-inflammatory cytokines and molecules. Thus, cGAS-STING signaling mediated recognition of cytosolic dsDNA causing the induction of type 1 IFNs plays a crucial role in innate immunity against cytosolic pathogens, PAMPs, and DAMPs. The overactivation of this system may lead to the development of autoinflammation and autoimmune diseases. The article opens with the introduction of different PRRs involved in the intracellular recognition of dsDNA and gives a brief introduction of cGAS-STING signaling. The second section briefly describes cGAS as intracellular PRR required to recognize intracellular nucleic acids (dsDNA and CDNs) and the formation of cGAMP. The cGAMP acts as a second messenger to activate STING- and TANK-binding kinase 1-mediated generation of type 1 IFNs and the activation of NF-κB. The third section of the article describes the role of cGAS-STING signaling in the induction of autoinflammation and various autoimmune diseases. The subsequent fourth section describes both chemical compounds developed and the endogenous negative regulators of cGAS-STING signaling required for its regulation. Therapeutic targeting of cGAS-STING signaling could offer new ways to treat inflammatory and autoimmune diseases.


Subject(s)
Autoimmune Diseases/etiology , Inflammation/etiology , Membrane Proteins/physiology , Animals , DNA/metabolism , Exodeoxyribonucleases/physiology , Extracellular Traps/physiology , Humans , Interferon Type I/physiology , Membrane Proteins/antagonists & inhibitors , Nucleotides, Cyclic/physiology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/physiology , Phosphoproteins/physiology , Signal Transduction/physiology
20.
Cell ; 176(6): 1432-1446.e11, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827685

ABSTRACT

The presence of DNA in the cytosol of mammalian cells is an unusual event that is often associated with genotoxic stress or viral infection. The enzyme cGAS is a sensor of cytosolic DNA that induces interferon and inflammatory responses that can be protective or pathologic, depending on the context. Along with other cytosolic innate immune receptors, cGAS is thought to diffuse throughout the cytosol in search of its DNA ligand. Herein, we report that cGAS is not a cytosolic protein but rather localizes to the plasma membrane via the actions of an N-terminal phosphoinositide-binding domain. This domain interacts selectively with PI(4,5)P2, and cGAS mutants defective for lipid binding are mislocalized to the cytosolic and nuclear compartments. Mislocalized cGAS induces potent interferon responses to genotoxic stress, but weaker responses to viral infection. These data establish the subcellular positioning of a cytosolic innate immune receptor as a mechanism that governs self-nonself discrimination.


Subject(s)
Cell Membrane/physiology , Nucleotidyltransferases/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cytosol/physiology , DNA, Viral/genetics , Female , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate/physiology , Interferons/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nucleotidyltransferases/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Phosphatidylinositols , Protein Binding , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...