Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 21(7): 727-735, 2020 07.
Article in English | MEDLINE | ID: mdl-32541831

ABSTRACT

Stimulator-of-interferon genes (STING) is vital for sensing cytosolic DNA and initiating innate immune responses against microbial infection and tumors. Redox homeostasis is the balance of oxidative and reducing reactions present in all living systems. Yet, how the intracellular redox state controls STING activation is unclear. Here, we show that cellular redox homeostasis maintained by glutathione peroxidase 4 (GPX4) is required for STING activation. GPX4 deficiency enhanced cellular lipid peroxidation and thus specifically inhibited the cGAS-STING pathway. Concordantly, GPX4 deficiency inhibited herpes simplex virus-1 (HSV-1)-induced innate antiviral immune responses and promoted HSV-1 replication in vivo. Mechanistically, GPX4 inactivation increased production of lipid peroxidation, which led to STING carbonylation at C88 and inhibited its trafficking from the endoplasmic reticulum (ER) to the Golgi complex. Thus, cellular stress-induced lipid peroxidation specifically attenuates the STING DNA-sensing pathway, suggesting that GPX4 facilitates STING activation by maintaining redox homeostasis of lipids.


Subject(s)
Herpes Simplex/immunology , Membrane Proteins/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Animals , Carbolines/pharmacology , Cells, Cultured , DNA, Viral/immunology , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Female , Fibroblasts , Golgi Apparatus/metabolism , HEK293 Cells , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Homeostasis/immunology , Humans , Immunity, Innate , Lipid Peroxidation/genetics , Lipid Peroxidation/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Membrane Proteins/immunology , Mice , Mice, Knockout , Nucleotidyltransferases/metabolism , Oxidation-Reduction , Oximes/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Primary Cell Culture , Protein Carbonylation/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Sulfonamides/pharmacology , THP-1 Cells , Virus Replication/immunology
3.
Immunity ; 54(9): 1961-1975.e5, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525337

ABSTRACT

Nucleic acids are powerful triggers of innate immunity and can adopt the Z-conformation, an unusual left-handed double helix. Here, we studied the biological function(s) of Z-RNA recognition by the adenosine deaminase ADAR1, mutations in which cause Aicardi-Goutières syndrome. Adar1mZα/mZα mice, bearing two point mutations in the Z-nucleic acid binding (Zα) domain that abolish Z-RNA binding, displayed spontaneous induction of type I interferons (IFNs) in multiple organs, including in the lung, where both stromal and hematopoietic cells showed IFN-stimulated gene (ISG) induction. Lung neutrophils expressed ISGs induced by the transcription factor IRF3, indicating an initiating role for neutrophils in this IFN response. The IFN response in Adar1mZα/mZα mice required the adaptor MAVS, implicating cytosolic RNA sensing. Adenosine-to-inosine changes were enriched in transposable elements and revealed a specific requirement of ADAR1's Zα domain in editing of a subset of RNAs. Thus, endogenous RNAs in Z-conformation have immunostimulatory potential curtailed by ADAR1, with relevance to autoinflammatory disease in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adenosine Deaminase/genetics , Interferon Type I/immunology , RNA, Double-Stranded/genetics , Adenosine/genetics , Adenosine/metabolism , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Inosine/genetics , Inosine/metabolism , Interferon Type I/genetics , Mice , Mutation , Nervous System Malformations/genetics , Nervous System Malformations/immunology , RNA Editing/genetics , RNA, Double-Stranded/metabolism
4.
Mol Cell ; 81(20): 4109-4110, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34686311

ABSTRACT

Unusual nucleic acids activate innate immunity and may be present in transformed cells. Meng et al. (2021) find that cancer-associated mutations in NF2 turn this tumor suppressor into a potent antagonist of DNA- and RNA-induced innate immune signaling.


Subject(s)
Interferons , Nucleic Acids , Immunity, Innate , RNA , Signal Transduction
5.
Proc Natl Acad Sci U S A ; 121(4): e2309628121, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38227660

ABSTRACT

Human bone marrow failure (BMF) syndromes result from the loss of hematopoietic stem and progenitor cells (HSPC), and this loss has been attributed to cell death; however, the cell death triggers, and mechanisms remain unknown. During BMF, tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ) increase. These ligands are known to induce necroptosis, an inflammatory form of cell death mediated by RIPK1, RIPK3, and MLKL. We previously discovered that mice with a hematopoietic RIPK1 deficiency (Ripk1HEM KO) exhibit inflammation, HSPC loss, and BMF, which is partially ameliorated by a RIPK3 deficiency; however, whether RIPK3 exerts its effects through its function in mediating necroptosis or other forms of cell death remains unclear. Here, we demonstrate that similar to a RIPK3 deficiency, an MLKL deficiency significantly extends survival and like Ripk3 deficiency partially restores hematopoiesis in Ripk1HEM KO mice revealing that both necroptosis and apoptosis contribute to BMF in these mice. Using mouse models, we show that the nucleic acid sensor Z-DNA binding protein 1 (ZBP1) is up-regulated in mouse RIPK1-deficient bone marrow cells and that ZBP1's function in endogenous nucleic acid sensing is necessary for HSPC death and contributes to BMF. We also provide evidence that IFNγ mediates HSPC death in Ripk1HEM KO mice, as ablation of IFNγ but not TNFα receptor signaling significantly extends survival of these mice. Together, these data suggest that RIPK1 maintains hematopoietic homeostasis by preventing ZBP1 activation and induction of HSPC death.


Subject(s)
Nucleic Acids , Pancytopenia , Animals , Humans , Mice , Apoptosis/genetics , Bone Marrow Failure Disorders , Cell Death/physiology , Hematopoietic Stem Cells/metabolism , Necrosis/metabolism , Nucleic Acids/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
6.
EMBO J ; 41(14): e109217, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35670106

ABSTRACT

Varicella-Zoster virus (VZV) causes chickenpox and shingles. Although the infection is associated with severe morbidity in some individuals, molecular mechanisms that determine innate immune responses remain poorly defined. We found that the cGAS/STING DNA sensing pathway was required for type I interferon (IFN) induction during VZV infection and that recognition of VZV by cGAS restricted its replication. Screening of a VZV ORF expression library identified the essential VZV tegument protein ORF9 as a cGAS antagonist. Ectopically or virally expressed ORF9 bound to endogenous cGAS leading to reduced type I IFN responses to transfected DNA. Confocal microscopy revealed co-localisation of cGAS and ORF9. ORF9 and cGAS also interacted directly in a cell-free system and phase-separated together with DNA. Furthermore, ORF9 inhibited cGAMP production by cGAS. Taken together, these results reveal the importance of the cGAS/STING DNA sensing pathway for VZV recognition and identify a VZV immune antagonist that partially but directly interferes with DNA sensing via cGAS.


Subject(s)
Herpesvirus 3, Human , Interferon Type I , Nucleotidyltransferases , Viral Proteins , DNA/metabolism , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/immunology , Humans , Immunity, Innate , Interferon Type I/immunology , Membrane Proteins/immunology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/immunology , Viral Proteins/immunology
7.
Immunity ; 46(3): 337-339, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329697

ABSTRACT

Detection of cyclic dinucleotides (cdNs) by the STING pathway potently triggers the antiviral response. McFarland et al. now show that the mouse oxidoreductase RECON acts as a sensor for some bacterial cdNs, modulating innate signaling in a manner independent of STING to promote an antibacterial state.


Subject(s)
Membrane Proteins , Signal Transduction , Animals , Mice
8.
Cell ; 140(3): 397-408, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20144762

ABSTRACT

RIG-I is a key mediator of antiviral immunity, able to couple detection of infection by RNA viruses to the induction of interferons. Natural RIG-I stimulatory RNAs have variously been proposed to correspond to virus genomes, virus replication intermediates, viral transcripts, or self-RNA cleaved by RNase L. However, the relative contribution of each of these RNA species to RIG-I activation and interferon induction in virus-infected cells is not known. Here, we use three approaches to identify physiological RIG-I agonists in cells infected with influenza A virus or Sendai virus. We show that RIG-I agonists are exclusively generated by the process of virus replication and correspond to full-length virus genomes. Therefore, nongenomic viral transcripts, short replication intermediates, and cleaved self-RNA do not contribute substantially to interferon induction in cells infected with these negative strand RNA viruses. Rather, single-stranded RNA viral genomes bearing 5'-triphosphates constitute the natural RIG-I agonists that trigger cell-intrinsic innate immune responses during infection.


Subject(s)
DEAD-box RNA Helicases/immunology , Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , RNA Virus Infections/immunology , RNA, Viral/immunology , Animals , Cell Line , DEAD Box Protein 58 , Dogs , Humans , Interferons/immunology , Mice , RNA Viruses/physiology , Receptors, Cell Surface , Receptors, Immunologic , Virus Replication
9.
Genes Dev ; 31(4): 353-369, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28279982

ABSTRACT

Radiotherapy and chemotherapy are effective treatment methods for many types of cancer, but resistance is common. Recent findings indicate that antiviral type I interferon (IFN) signaling is induced by these treatments. However, the underlying mechanisms still need to be elucidated. Expression of a set of IFN-stimulated genes comprises an IFN-related DNA damage resistance signature (IRDS), which correlates strongly with resistance to radiotherapy and chemotherapy across different tumors. Classically, during viral infection, the presence of foreign DNA in the cytoplasm of host cells can initiate type I IFN signaling. Here, we demonstrate that DNA-damaging modalities used during cancer therapy lead to the release of ssDNA fragments from the cell nucleus into the cytosol, engaging this innate immune response. We found that the factors that control DNA end resection during double-strand break repair, including the Bloom syndrome (BLM) helicase and exonuclease 1 (EXO1), play a major role in generating these DNA fragments and that the cytoplasmic 3'-5' exonuclease Trex1 is required for their degradation. Analysis of mRNA expression profiles in breast tumors demonstrates that those with lower Trex1 and higher BLM and EXO1 expression levels are associated with poor prognosis. Targeting BLM and EXO1 could therefore represent a novel approach for circumventing the IRDS produced in response to cancer therapeutics.


Subject(s)
DNA Damage , Exodeoxyribonucleases/metabolism , Immunity, Innate/genetics , Interferons/metabolism , Phosphoproteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Cell Line, Tumor , Cytoplasm/enzymology , Cytoplasm/immunology , Cytoplasm/metabolism , DNA Damage/drug effects , DNA, Single-Stranded/immunology , DNA, Single-Stranded/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mutagens/therapeutic use , Mutagens/toxicity , Radiation Tolerance/immunology , Radiation, Ionizing , Reactive Oxygen Species , Signal Transduction
10.
EMBO J ; 39(15): e102931, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32511795

ABSTRACT

Sterile alpha motif and histidine-aspartic acid domain-containing protein 1 (SAMHD1), a dNTP triphosphohydrolase, regulates the levels of cellular dNTPs through their hydrolysis. SAMHD1 protects cells from invading viruses that depend on dNTPs to replicate and is frequently mutated in cancers and Aicardi-Goutières syndrome, a hereditary autoimmune encephalopathy. We discovered that SAMHD1 localizes at the immunoglobulin (Ig) switch region, and serves as a novel DNA repair regulator of Ig class switch recombination (CSR). Depletion of SAMHD1 impaired not only CSR but also IgH/c-Myc translocation. Consistently, we could inhibit these two processes by elevating the cellular nucleotide pool. A high frequency of nucleotide insertion at the break-point junctions is a notable feature in SAMHD1 deficiency during activation-induced cytidine deaminase-mediated genomic instability. Interestingly, CSR induced by staggered but not blunt, double-stranded DNA breaks was impaired by SAMHD1 depletion, which was accompanied by enhanced nucleotide insertions at recombination junctions. We propose that SAMHD1-mediated dNTP balance regulates dNTP-sensitive DNA end-processing enzyme and promotes CSR and aberrant genomic rearrangements by suppressing the insertional DNA repair pathway.


Subject(s)
DNA Repair , Deoxyribonucleotides/metabolism , Immunoglobulin Class Switching , SAM Domain and HD Domain-Containing Protein 1/metabolism , Cell Line , Deoxyribonucleotides/genetics , Humans , SAM Domain and HD Domain-Containing Protein 1/genetics
11.
EMBO Rep ; 23(12): e55839, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36268590

ABSTRACT

ZBP1 is an interferon-induced cytosolic nucleic acid sensor that facilitates antiviral responses via RIPK3. Although ZBP1-mediated programmed cell death is widely described, whether and how it promotes inflammatory signaling is unclear. Here, we report a ZBP1-induced inflammatory signaling pathway mediated by K63- and M1-linked ubiquitin chains, which depends on RIPK1 and RIPK3 as scaffolds independently of cell death. In human HT29 cells, ZBP1 associated with RIPK1 and RIPK3 as well as ubiquitin ligases cIAP1 and LUBAC. ZBP1-induced K63- and M1-linked ubiquitination of RIPK1 and ZBP1 to promote TAK1- and IKK-mediated inflammatory signaling and cytokine production. Inhibition of caspase activity suppressed ZBP1-induced cell death but enhanced cytokine production in a RIPK1- and RIPK3 kinase activity-dependent manner. Lastly, we provide evidence that ZBP1 signaling contributes to SARS-CoV-2-induced cytokine production. Taken together, we describe a ZBP1-RIPK3-RIPK1-mediated inflammatory signaling pathway relayed by the scaffolding role of RIPKs and regulated by caspases, which may induce inflammation when ZBP1 is activated below the threshold needed to trigger a cell death response.


Subject(s)
Cell Death , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases , Humans , Cytokines , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Ubiquitin , RNA-Binding Proteins/genetics , HT29 Cells , Inflammation
12.
Nature ; 560(7717): 238-242, 2018 08.
Article in English | MEDLINE | ID: mdl-30046113

ABSTRACT

Mitochondria are descendants of endosymbiotic bacteria and retain essential prokaryotic features such as a compact circular genome. Consequently, in mammals, mitochondrial DNA is subjected to bidirectional transcription that generates overlapping transcripts, which are capable of forming long double-stranded RNA structures1,2. However, to our knowledge, mitochondrial double-stranded RNA has not been previously characterized in vivo. Here we describe the presence of a highly unstable native mitochondrial double-stranded RNA species at single-cell level and identify key roles for the degradosome components mitochondrial RNA helicase SUV3 and polynucleotide phosphorylase PNPase in restricting the levels of mitochondrial double-stranded RNA. Loss of either enzyme results in massive accumulation of mitochondrial double-stranded RNA that escapes into the cytoplasm in a PNPase-dependent manner. This process engages an MDA5-driven antiviral signalling pathway that triggers a type I interferon response. Consistent with these data, patients carrying hypomorphic mutations in the gene PNPT1, which encodes PNPase, display mitochondrial double-stranded RNA accumulation coupled with upregulation of interferon-stimulated genes and other markers of immune activation. The localization of PNPase to the mitochondrial inter-membrane space and matrix suggests that it has a dual role in preventing the formation and release of mitochondrial double-stranded RNA into the cytoplasm. This in turn prevents the activation of potent innate immune defence mechanisms that have evolved to protect vertebrates against microbial and viral attack.


Subject(s)
Herpesvirus 1, Human/immunology , RNA, Double-Stranded/immunology , RNA, Mitochondrial/immunology , Animals , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Exoribonucleases/deficiency , Exoribonucleases/genetics , Exoribonucleases/metabolism , Gene Expression Regulation/immunology , HeLa Cells , Herpesvirus 1, Human/genetics , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Mice , Mice, Inbred C57BL , Multienzyme Complexes/metabolism , Mutation , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , Single-Cell Analysis , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
13.
Ann Rheum Dis ; 82(6): 754-762, 2023 06.
Article in English | MEDLINE | ID: mdl-36858821

ABSTRACT

BACKGROUND: Type I interferons (IFN-Is) play a role in a broad range of rheumatic and musculoskeletal diseases (RMDs), and compelling evidence suggests that their measurement could have clinical value, although testing has not progressed into clinical settings. OBJECTIVE: To develop evidence-based points to consider (PtC) for the measurement and reporting of IFN-I assays in clinical research and to determine their potential clinical utility. METHODS: EULAR standardised operating procedures were followed. A task force including rheumatologists, immunologists, translational scientists and a patient partner was formed. Two systematic reviews were conducted to address methodological and clinical questions. PtC were formulated based on the retrieved evidence and expert opinion. Level of evidence and agreement was determined. RESULTS: Two overarching principles and 11 PtC were defined. The first set (PtC 1-4) concerned terminology, assay characteristics and reporting practices to enable more consistent reporting and facilitate translation and collaborations. The second set (PtC 5-11) addressed clinical applications for diagnosis and outcome assessments, including disease activity, prognosis and prediction of treatment response. The mean level of agreement was generally high, mainly in the first PtC set and for clinical applications in systemic lupus erythematosus. Harmonisation of assay methodology and clinical validation were key points for the research agenda. CONCLUSIONS: IFN-I assays have a high potential for implementation in the clinical management of RMDs. Uptake of these PtC will facilitate the progress of IFN-I assays into clinical practice and may be also of interest beyond rheumatology.


Subject(s)
Musculoskeletal Diseases , Rheumatology , Humans
14.
EMBO Rep ; 22(8): e52447, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34142428

ABSTRACT

Cyclic GMP-AMP (cGAMP) is an immunostimulatory molecule produced by cGAS that activates STING. cGAMP is an adjuvant when administered alongside antigens. cGAMP is also incorporated into enveloped virus particles during budding. Here, we investigate whether inclusion of cGAMP within viral vaccine vectors enhances their immunogenicity. We immunise mice with virus-like particles (VLPs) containing HIV-1 Gag and the vesicular stomatitis virus envelope glycoprotein G (VSV-G). cGAMP loading of VLPs augments CD4 and CD8 T-cell responses. It also increases VLP- and VSV-G-specific antibody titres in a STING-dependent manner and enhances virus neutralisation, accompanied by increased numbers of T follicular helper cells. Vaccination with cGAMP-loaded VLPs containing haemagglutinin induces high titres of influenza A virus neutralising antibodies and confers protection upon virus challenge. This requires cGAMP inclusion within VLPs and is achieved at markedly reduced cGAMP doses. Similarly, cGAMP loading of VLPs containing the SARS-CoV-2 Spike protein enhances Spike-specific antibody titres. cGAMP-loaded VLPs are thus an attractive platform for vaccination.


Subject(s)
COVID-19 , Influenza Vaccines , Vaccines, Virus-Like Particle , Animals , Humans , Mice , Nucleotides, Cyclic , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccines, Virus-Like Particle/genetics
15.
EMBO Rep ; 21(12): e51345, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33155371

ABSTRACT

Cell-autonomous sensing of nucleic acids is essential for host defence against invading pathogens by inducing antiviral and inflammatory cytokines. cGAS has emerged in recent years as a non-redundant DNA sensor important for detection of many viruses and bacteria. Upon binding to DNA, cGAS synthesises the cyclic dinucleotide 2'3'-cGAMP that binds to the adaptor protein STING and thereby triggers IRF3- and NFκB-dependent transcription. In addition to infection, the pathophysiology of an ever-increasing number of sterile inflammatory conditions in humans involves the recognition of DNA through cGAS. Consequently, the cGAS/STING signalling axis has emerged as an attractive target for pharmacological modulation. However, the development of cGAS and STING inhibitors has just begun and a need for specific and effective compounds persists. In this review, we focus on cGAS and explore how its activation by immunostimulatory DNA is regulated by cellular mechanisms, viral immune modulators and small molecules. We further use our knowledge of cGAS modulation by cells and viruses to conceptualise potential new ways of pharmacological cGAS targeting.


Subject(s)
Membrane Proteins , Nucleotidyltransferases , Cytokines , DNA/genetics , Humans , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction
16.
EMBO J ; 36(17): 2529-2543, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28716805

ABSTRACT

Nucleic acids are potent triggers for innate immunity. Double-stranded DNA and RNA adopt different helical conformations, including the unusual Z-conformation. Z-DNA/RNA is recognised by Z-binding domains (ZBDs), which are present in proteins implicated in antiviral immunity. These include ZBP1 (also known as DAI or DLM-1), which induces necroptosis, an inflammatory form of cell death. Using reconstitution and knock-in models, we report that mutation of key amino acids involved in Z-DNA/RNA binding in ZBP1's ZBDs prevented necroptosis upon infection with mouse cytomegalovirus. Induction of cell death was cell autonomous and required RNA synthesis but not viral DNA replication. Accordingly, ZBP1 directly bound to RNA via its ZBDs. Intact ZBP1-ZBDs were also required for necroptosis triggered by ectopic expression of ZBP1 and caspase blockade, and ZBP1 cross-linked to endogenous RNA These observations show that Z-RNA may constitute a molecular pattern that induces inflammatory cell death upon sensing by ZBP1.


Subject(s)
Apoptosis/physiology , Glycoproteins/metabolism , RNA/metabolism , Animals , Cytomegalovirus/genetics , Cytomegalovirus Infections/metabolism , Glycoproteins/genetics , Mice , Mice, Transgenic , NIH 3T3 Cells , Nucleic Acid Conformation , RNA/chemistry , RNA-Binding Proteins
17.
EMBO J ; 36(5): 604-616, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28122869

ABSTRACT

An unresolved question is how HIV-1 achieves efficient replication in terminally differentiated macrophages despite the restriction factor SAMHD1. We reveal inducible changes in expression of cell cycle-associated proteins including MCM2 and cyclins A, E, D1/D3 in macrophages, without evidence for DNA synthesis or mitosis. These changes are induced by activation of the Raf/MEK/ERK kinase cascade, culminating in upregulation of CDK1 with subsequent SAMHD1 T592 phosphorylation and deactivation of its antiviral activity. HIV infection is limited to these G1-like phase macrophages at the single-cell level. Depletion of SAMHD1 in macrophages decouples the association between infection and expression of cell cycle-associated proteins, with terminally differentiated macrophages becoming highly susceptible to HIV-1. We observe both embryo-derived and monocyte-derived tissue-resident macrophages in a G1-like phase at frequencies approaching 20%, suggesting how macrophages sustain HIV-1 replication in vivo Finally, we reveal a SAMHD1-dependent antiretroviral activity of histone deacetylase inhibitors acting via p53 activation. These data provide a basis for host-directed therapeutic approaches aimed at limiting HIV-1 burden in macrophages that may contribute to curative interventions.


Subject(s)
G1 Phase , HIV-1/physiology , Immune Evasion , Macrophages/immunology , Macrophages/virology , Monomeric GTP-Binding Proteins/metabolism , Protein Processing, Post-Translational , Cells, Cultured , HIV-1/immunology , Humans , Immunity, Innate , Phosphorylation , SAM Domain and HD Domain-Containing Protein 1
18.
Eur J Immunol ; 50(1): 56-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31608988

ABSTRACT

Toll-like receptor 7 (TLR7) is an innate immune sensor for single-strand RNA (ssRNA). Recent structural analysis revealed that TLR7 has an additional binding site for nucleosides such as guanosine, and is activated when both guanosine and ssRNA bind. The nucleoside binding site also accommodates imidazoquinoline derivatives such as R848, which activate TLR7 in the absence of ssRNA. Here, we report that deoxyguanosine (dG) triggered cytokine production in murine bone marrow derived macrophages and plasmacytoid dendritic cells, as well as in human peripheral blood mononuclear cells, including type I interferons and pro-inflammatory factors such as TNF and IL-6. This signalling activity of dG was dependent on TLR7 and its adaptor MyD88 and did not require amplification via the type I interferon receptor. dG-triggered cytokine production required endosomal maturation but did not depend on the concurrent provision of RNA. We conclude that dG induces an inflammatory response through TLR7 and propose that dG is an RNA-independent TLR7 agonist.


Subject(s)
Deoxyguanosine/immunology , Inflammation/immunology , Toll-Like Receptor 7/agonists , Animals , Deoxyguanosine/metabolism , Humans , Inflammation/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL
19.
Proc Natl Acad Sci U S A ; 115(33): E7768-E7775, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061387

ABSTRACT

The adaptor molecule stimulator of IFN genes (STING) is central to production of type I IFNs in response to infection with DNA viruses and to presence of host DNA in the cytosol. Excessive release of type I IFNs through STING-dependent mechanisms has emerged as a central driver of several interferonopathies, including systemic lupus erythematosus (SLE), Aicardi-Goutières syndrome (AGS), and stimulator of IFN genes-associated vasculopathy with onset in infancy (SAVI). The involvement of STING in these diseases points to an unmet need for the development of agents that inhibit STING signaling. Here, we report that endogenously formed nitro-fatty acids can covalently modify STING by nitro-alkylation. These nitro-alkylations inhibit STING palmitoylation, STING signaling, and subsequently, the release of type I IFN in both human and murine cells. Furthermore, treatment with nitro-fatty acids was sufficient to inhibit production of type I IFN in fibroblasts derived from SAVI patients with a gain-of-function mutation in STING. In conclusion, we have identified nitro-fatty acids as endogenously formed inhibitors of STING signaling and propose for these lipids to be considered in the treatment of STING-dependent inflammatory diseases.


Subject(s)
Fatty Acids/metabolism , Herpes Simplex/metabolism , Herpesvirus 2, Human/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Herpes Simplex/genetics , Herpes Simplex/pathology , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Lipoylation , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , RAW 264.7 Cells
20.
Nature ; 514(7522): 372-375, 2014 10 16.
Article in English | MEDLINE | ID: mdl-25119032

ABSTRACT

Mammalian cells possess mechanisms to detect and defend themselves from invading viruses. In the cytosol, the RIG-I-like receptors (RLRs), RIG-I (retinoic acid-inducible gene I; encoded by DDX58) and MDA5 (melanoma differentiation-associated gene 5; encoded by IFIH1) sense atypical RNAs associated with virus infection. Detection triggers a signalling cascade via the adaptor MAVS that culminates in the production of type I interferons (IFN-α and ß; hereafter IFN), which are key antiviral cytokines. RIG-I and MDA5 are activated by distinct viral RNA structures and much evidence indicates that RIG-I responds to RNAs bearing a triphosphate (ppp) moiety in conjunction with a blunt-ended, base-paired region at the 5'-end (reviewed in refs 1, 2, 3). Here we show that RIG-I also mediates antiviral responses to RNAs bearing 5'-diphosphates (5'pp). Genomes from mammalian reoviruses with 5'pp termini, 5'pp-RNA isolated from yeast L-A virus, and base-paired 5'pp-RNAs made by in vitro transcription or chemical synthesis, all bind to RIG-I and serve as RIG-I agonists. Furthermore, a RIG-I-dependent response to 5'pp-RNA is essential for controlling reovirus infection in cultured cells and in mice. Thus, the minimal determinant for RIG-I recognition is a base-paired RNA with 5'pp. Such RNAs are found in some viruses but not in uninfected cells, indicating that recognition of 5'pp-RNA, like that of 5'ppp-RNA, acts as a powerful means of self/non-self discrimination by the innate immune system.


Subject(s)
DEAD-box RNA Helicases/metabolism , Diphosphates/metabolism , Immunity, Innate , RNA, Viral/chemistry , RNA, Viral/metabolism , Reoviridae/genetics , Reoviridae/immunology , Animals , Base Pairing , Base Sequence , DEAD Box Protein 58 , Female , Genome, Viral/genetics , Male , Mice , RNA, Viral/genetics , Reoviridae/physiology
SELECTION OF CITATIONS
SEARCH DETAIL