Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30270045

ABSTRACT

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Subject(s)
Capsid Proteins/immunology , Nuclear Matrix-Associated Proteins/immunology , Nuclear Matrix-Associated Proteins/physiology , Octamer Transcription Factors/immunology , Octamer Transcription Factors/physiology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/physiology , Capsid/metabolism , Capsid Proteins/metabolism , Capsid Proteins/physiology , Cell Nucleus/metabolism , DNA, Viral/genetics , DNA, Viral/immunology , DNA-Binding Proteins , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , HIV-2/genetics , HIV-2/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate/immunology , Macrophages/immunology , Membrane Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/physiology , RNA-Binding Proteins/metabolism , Signal Transduction/immunology
3.
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
4.
Nat Rev Mol Cell Biol ; 13(3): 141-52, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22358330

ABSTRACT

The past 15 years have seen an explosion in our understanding of how cells replicate damaged DNA and how this can lead to mutagenesis. The Y-family DNA polymerases lie at the heart of this process, which is commonly known as translesion synthesis. This family of polymerases has unique features that enable them to synthesize DNA past damaged bases. However, as they exhibit low fidelity when copying undamaged DNA, it is essential that they are only called into play when they are absolutely required. Several layers of regulation ensure that this is achieved.


Subject(s)
DNA Damage , Nucleotidyltransferases/physiology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Catalytic Domain , DNA Repair , DNA Replication , Humans , Mutagenesis , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Protein Binding , Protein Structure, Tertiary
5.
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
6.
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
7.
EMBO J ; 37(8)2018 04 13.
Article in English | MEDLINE | ID: mdl-29496741

ABSTRACT

Negative regulation of immune pathways is essential to achieve resolution of immune responses and to avoid excess inflammation. DNA stimulates type I IFN expression through the DNA sensor cGAS, the second messenger cGAMP, and the adaptor molecule STING Here, we report that STING degradation following activation of the pathway occurs through autophagy and is mediated by p62/SQSTM1, which is phosphorylated by TBK1 to direct ubiquitinated STING to autophagosomes. Degradation of STING was impaired in p62-deficient cells, which responded with elevated IFN production to foreign DNA and DNA pathogens. In the absence of p62, STING failed to traffic to autophagy-associated vesicles. Thus, DNA sensing induces the cGAS-STING pathway to activate TBK1, which phosphorylates IRF3 to induce IFN expression, but also phosphorylates p62 to stimulate STING degradation and attenuation of the response.


Subject(s)
Nucleotidyltransferases/physiology , Protein Serine-Threonine Kinases/physiology , Sequestosome-1 Protein/physiology , Animals , Autophagy , Cell Line , DNA/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
8.
Mol Cell ; 54(2): 289-96, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766893

ABSTRACT

The innate immune system deploys a variety of sensors to detect signs of infection. Nucleic acids represent a major class of pathogen signatures that can trigger robust immune responses. The presence of DNA in the cytoplasm of mammalian cells is a danger signal that activates innate immune responses; however, how cytosolic DNA triggers these responses remained unclear until recently. In this review, we focus on the mechanism of DNA sensing by the newly discovered cGAS-cGAMP-STING pathway and highlight recent progress in dissecting the in vivo functions of this pathway in immune defense as well as autoimmunity.


Subject(s)
Cytosol/metabolism , DNA/metabolism , Immunity, Innate/genetics , Models, Immunological , Nucleotides, Cyclic/physiology , Nucleotidyltransferases/physiology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Humans , Membrane Proteins/metabolism , Membrane Proteins/physiology , Models, Molecular , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction
9.
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
10.
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
11.
PLoS Pathog ; 14(1): e1006829, 2018 01.
Article in English | MEDLINE | ID: mdl-29298342

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-STING pathway is central for innate immune sensing of various bacterial, viral and protozoal infections. Recent studies identified the common HAQ and R232H alleles of TMEM173/STING, but the functional consequences of these variants for primary infections are unknown. Here we demonstrate that cGAS- and STING-deficient murine macrophages as well as human cells of individuals carrying HAQ TMEM173/STING were severely impaired in producing type I IFNs and pro-inflammatory cytokines in response to Legionella pneumophila, bacterial DNA or cyclic dinucleotides (CDNs). In contrast, R232H attenuated cytokine production only following stimulation with bacterial CDN, but not in response to L. pneumophila or DNA. In a mouse model of Legionnaires' disease, cGAS- and STING-deficient animals exhibited higher bacterial loads as compared to wild-type mice. Moreover, the haplotype frequency of HAQ TMEM173/STING, but not of R232H TMEM173/STING, was increased in two independent cohorts of human Legionnaires' disease patients as compared to healthy controls. Our study reveals that the cGAS-STING cascade contributes to antibacterial defense against L. pneumophila in mice and men, and provides important insight into how the common HAQ TMEM173/STING variant affects antimicrobial immune responses and susceptibility to infection. TRIAL REGISTRATION: ClinicalTrials.gov DRKS00005274, German Clinical Trials Register.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Immunity, Innate/genetics , Legionella pneumophila/immunology , Legionnaires' Disease/drug therapy , Legionnaires' Disease/genetics , Membrane Proteins/genetics , Nucleotidyltransferases/physiology , Adult , Aged , Aged, 80 and over , Animals , Case-Control Studies , Cells, Cultured , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Immunity, Innate/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Polymorphism, Genetic , Treatment Outcome
12.
PLoS Pathog ; 14(4): e1006976, 2018 04.
Article in English | MEDLINE | ID: mdl-29608601

ABSTRACT

In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2'3'-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2'3'-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2'3'-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo.


Subject(s)
Antiviral Agents/pharmacology , Herpes Genitalis/prevention & control , Herpesvirus 2, Human/drug effects , Host-Pathogen Interactions/drug effects , Membrane Proteins/agonists , Nucleotides, Cyclic/pharmacology , Animals , Cells, Cultured , Cytosol/virology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Herpes Genitalis/metabolism , Herpes Genitalis/virology , Herpesvirus 2, Human/pathogenicity , Humans , Interferon Type I/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/virology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Nucleotidyltransferases/physiology , Signal Transduction
13.
Mol Cell Proteomics ; 17(4): 607-618, 2018 04.
Article in English | MEDLINE | ID: mdl-29371291

ABSTRACT

The systematic investigation of gene mutation and expression is important to discover novel biomarkers and therapeutic targets in cancers. Here, we integrated genomics, transcriptomics, proteomics, and metabolomics to analyze three hepatocellular carcinoma (HCC) cell lines with differential metastatic potentials. The results revealed the profile of the prometastasis metabolism potentially associated with HCC metastasis. The multiomic analysis identified 12 genes with variations at multiple levels from three metabolic pathways, including glycolysis, starch, and sucrose metabolism, and glutathione metabolism. Furthermore, uridine diphosphate (UDP)-glucose pyrophosphorylase 2 (UGP2), was observed to be persistently up-regulated with increased metastatic potential. UGP2 overexpression promoted cell migration and invasion and enhanced glycogenesis in vitro The role of UGP2 in metastasis was further confirmed using a tumor xenograft mouse model. Taken together, the compendium of multiomic data provides valuable insights in understanding the roles of shifted cellular metabolism in HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genomics , Glucose/metabolism , Glycolysis , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Metabolomics , Neoplasm Invasiveness/genetics , Nucleotidyltransferases/physiology , Proteomics , Starch/metabolism
14.
Mol Cell ; 41(1): 117-27, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21211727

ABSTRACT

Bacteria regulate the assimilation of multiple nutrients to enable growth. How is balanced utilization achieved, despite fluctuations in the concentrations of the enzymes that make up the regulatory circuitry? Here we address this question by studying the nitrogen system of E. coli. A mechanism based on the avidity of a bifunctional enzyme, adenylyltransferase (AT/AR), to its multimeric substrate, glutamine synthetase, is proposed to maintain a robust ratio between two key metabolites, glutamine and α-ketoglutarate. This ratio is predicted to be insensitive to variations in protein levels of the core circuit and to the rate of nitrogen utilization. We find using mass spectrometry that the metabolite ratio is robust to variations in protein levels and that this robustness depends on the bifunctional enzyme. Moreover, robustness carries through to the bacteria growth rate. Interrupting avidity by adding a monofunctional AT/AR mutant to the native system abolishes robustness, as predicted by the proposed mechanism.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/enzymology , Nitrogen/metabolism , Nucleotidyltransferases/physiology , Carbon/metabolism , Escherichia coli/growth & development , Escherichia coli Proteins/metabolism , Glutamate-Ammonia Ligase/metabolism , Mass Spectrometry , Models, Biological , Protein Multimerization
15.
Nucleic Acids Res ; 44(22): 10824-10833, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27683219

ABSTRACT

Rev1 is a member of the Y-family of DNA polymerases and is known for its deoxycytidyl transferase activity that incorporates dCMP into DNA and its ability to function as a scaffold factor for other Y-family polymerases in translesion bypass events. Rev1 also is involved in mutagenic processes during somatic hypermutation of immunoglobulin genes. In light of the mutation pattern consistent with dCMP insertion observed earlier in mouse fibroblast cells treated with a base excision repair-inducing agent, we questioned whether Rev1 could also be involved in base excision repair (BER). Here, we uncovered a weak 5'-deoxyribose phosphate (5'-dRP) lyase activity in mouse Rev1 and demonstrated the enzyme can mediate BER in vitro The full-length Rev1 protein and its catalytic core domain are similar in their ability to support BER in vitro The dRP lyase activity in both of these proteins was confirmed by NaBH4 reduction of the Schiff base intermediate and kinetics studies. Limited proteolysis, mass spectrometry and deletion analysis localized the dRP lyase active site to the C-terminal segment of Rev1's catalytic core domain. These results suggest that Rev1 could serve as a backup polymerase in BER and could potentially contribute to AID-initiated antibody diversification through this activity.


Subject(s)
DNA Repair , Nucleotidyltransferases/physiology , Amino Acid Sequence , Animals , Catalytic Domain , DNA-Directed DNA Polymerase , Kinetics , Mice , Nucleotidyltransferases/chemistry , Peptide Mapping , Phosphorus-Oxygen Lyases/chemistry , Phosphorus-Oxygen Lyases/physiology , Proteolysis
16.
RNA ; 21(2): 243-53, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505023

ABSTRACT

The identity of tRNA(His) is strongly associated with the presence of an additional 5'-guanosine residue (G-1) in all three domains of life. The critical nature of the G-1 residue is underscored by the fact that two entirely distinct mechanisms for its acquisition are observed, with cotranscriptional incorporation observed in Bacteria, while post-transcriptional addition of G-1 occurs in Eukarya. Here, through our investigation of eukaryotes that lack obvious homologs of the post-transcriptional G-1-addition enzyme Thg1, we identify alternative pathways to tRNA(His) identity that controvert these well-established rules. We demonstrate that Trypanosoma brucei, like Acanthamoeba castellanii, lacks the G-1 identity element on tRNA(His) and utilizes a noncanonical G-1-independent histidyl-tRNA synthetase (HisRS). Purified HisRS enzymes from A. castellanii and T. brucei exhibit a mechanism of tRNA(His) recognition that is distinct from canonical G-1-dependent synthetases. Moreover, noncanonical HisRS enzymes genetically complement the loss of THG1 in Saccharomyces cerevisiae, demonstrating the biological relevance of the G-1-independent aminoacylation activity. In contrast, in Caenorhabditis elegans, which is another Thg1-independent eukaryote, the G-1 residue is maintained, but here its acquisition is noncanonical. In this case, the G-1 is encoded and apparently retained after 5' end processing, which has so far only been observed in Bacteria and organelles. Collectively, these observations unearth a widespread and previously unappreciated diversity in eukaryotic tRNA(His) identity mechanisms.


Subject(s)
Acanthamoeba castellanii/enzymology , RNA Processing, Post-Transcriptional , RNA, Protozoan/biosynthesis , RNA, Transfer, His/biosynthesis , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans/genetics , Histidine-tRNA Ligase/chemistry , Histidine-tRNA Ligase/physiology , Kinetics , Molecular Sequence Data , Nucleotidyltransferases/physiology , Phylogeny , Protozoan Proteins/chemistry , Protozoan Proteins/physiology , RNA, Protozoan/genetics , RNA, Transfer, His/genetics , Saccharomyces cerevisiae/genetics , Transfer RNA Aminoacylation
17.
PLoS Pathog ; 11(10): e1005253, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26506431

ABSTRACT

Encapsidation is a strategy almost universally employed by viruses to protect their genomes from degradation and from innate immune sensors. We show that TRIM21, which targets antibody-opsonized virions for proteasomal destruction, circumvents this protection, enabling the rapid detection and degradation of viral genomes before their replication. TRIM21 triggers an initial wave of cytokine transcription that is antibody, rather than pathogen, driven. This early response is augmented by a second transcriptional program, determined by the nature of the infecting virus. In this second response, TRIM21-induced exposure of the viral genome promotes sensing of DNA and RNA viruses by cGAS and RIG-I. This mechanism allows early detection of an infection event and drives an inflammatory response in mice within hours of viral challenge.


Subject(s)
DEAD-box RNA Helicases/physiology , Genome, Viral , Nucleotidyltransferases/physiology , Phagocytosis , Ribonucleoproteins/physiology , Virus Diseases/immunology , Adenovirus Infections, Human/immunology , Animals , DEAD Box Protein 58 , HeLa Cells , Humans , Immunity, Innate , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Picornaviridae Infections/immunology , Receptors, Immunologic , Rhinovirus
18.
Nucleic Acids Res ; 43(17): 8325-39, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26187992

ABSTRACT

REV1 is a eukaryotic member of the Y-family of DNA polymerases involved in translesion DNA synthesis and genome mutagenesis. Recently, REV1 is also found to function in homologous recombination. However, it remains unclear how REV1 is recruited to the sites where homologous recombination is processed. Here, we report that loss of mammalian REV1 results in a specific defect in replication-associated gene conversion. We found that REV1 is targeted to laser-induced DNA damage stripes in a manner dependent on its ubiquitin-binding motifs, on RAD18, and on monoubiquitinated FANCD2 (FANCD2-mUb) that associates with REV1. Expression of a FANCD2-Ub chimeric protein in RAD18-depleted cells enhances REV1 assembly at laser-damaged sites, suggesting that FANCD2-mUb functions downstream of RAD18 to recruit REV1 to DNA breaks. Consistent with this suggestion we found that REV1 and FANCD2 are epistatic with respect to sensitivity to the double-strand break-inducer camptothecin. REV1 enrichment at DNA damage stripes also partially depends on BRCA1 and BRCA2, components of the FANCD2/BRCA supercomplex. Intriguingly, analogous to FANCD2-mUb and BRCA1/BRCA2, REV1 plays an unexpected role in protecting nascent replication tracts from degradation by stabilizing RAD51 filaments. Collectively these data suggest that REV1 plays multiple roles at stalled replication forks in response to replication stress.


Subject(s)
DNA Damage , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/physiology , Nuclear Proteins/physiology , Nucleotidyltransferases/physiology , Camptothecin/toxicity , Cell Line , DNA/metabolism , DNA-Binding Proteins/physiology , DNA-Directed DNA Polymerase , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Conversion , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Protein Interaction Domains and Motifs , Stress, Physiological/genetics , Topoisomerase I Inhibitors/toxicity , Ubiquitin-Protein Ligases
19.
J Biol Chem ; 290(13): 8482-99, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25601083

ABSTRACT

The maintenance of endoplasmic reticulum (ER) homeostasis is a critical aspect of determining cell fate and requires a properly functioning unfolded protein response (UPR). We have discovered a previously unknown role of a post-translational modification termed adenylylation/AMPylation in regulating signal transduction events during UPR induction. A family of enzymes, defined by the presence of a Fic (filamentation induced by cAMP) domain, catalyzes this adenylylation reaction. The human genome encodes a single Fic protein, called HYPE (Huntingtin yeast interacting protein E), with adenylyltransferase activity but unknown physiological target(s). Here, we demonstrate that HYPE localizes to the lumen of the endoplasmic reticulum via its hydrophobic N terminus and adenylylates the ER molecular chaperone, BiP, at Ser-365 and Thr-366. BiP functions as a sentinel for protein misfolding and maintains ER homeostasis. We found that adenylylation enhances BiP's ATPase activity, which is required for refolding misfolded proteins while coping with ER stress. Accordingly, HYPE expression levels increase upon stress. Furthermore, siRNA-mediated knockdown of HYPE prevents the induction of an unfolded protein response. Thus, we identify HYPE as a new UPR regulator and provide the first functional data for Fic-mediated adenylylation in mammalian signaling.


Subject(s)
Carrier Proteins/physiology , Membrane Proteins/physiology , Nucleotidyltransferases/physiology , Protein Processing, Post-Translational , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Apoptosis , Cell Survival , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Glycosylation , HEK293 Cells , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Protein Structure, Tertiary , Protein Transport , Signal Transduction , Up-Regulation , eIF-2 Kinase/metabolism
20.
Biochim Biophys Acta ; 1847(9): 798-808, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25626174

ABSTRACT

During the evolution of eukaryotic genomes, many genes have been interrupted by intervening sequences (introns) that must be removed post-transcriptionally from RNA precursors to form mRNAs ready for translation. The origin of nuclear introns is still under debate, but one hypothesis is that the spliceosome and the intron-exon structure of genes have evolved from bacterial-type group II introns that invaded the eukaryotic genomes. The group II introns were most likely introduced into the eukaryotic genome from an α-proteobacterial predecessor of mitochondria early during the endosymbiosis event. These self-splicing and mobile introns spread through the eukaryotic genome and later degenerated. Pieces of introns became part of the general splicing machinery we know today as the spliceosome. In addition, group II introns likely brought intron maturases with them to the nucleus. Maturases are found in most bacterial introns, where they act as highly specific splicing factors for group II introns. In the spliceosome, the core protein Prp8 shows homology to group II intron-encoded maturases. While maturases are entirely intron specific, their descendant of the spliceosomal machinery, the Prp8 protein, is an extremely versatile splicing factor with multiple interacting proteins and RNAs. How could such a general player in spliceosomal splicing evolve from the monospecific bacterial maturases? Analysis of the organellar splicing machinery in plants may give clues on the evolution of nuclear splicing. Plants encode various proteins which are closely related to bacterial maturases. The organellar genomes contain one maturase each, named MatK in chloroplasts and MatR in mitochondria. In addition, several maturase genes have been found in the nucleus as well, which are acting on mitochondrial pre-RNAs. All plant maturases show sequence deviation from their progenitor bacterial maturases, and interestingly are all acting on multiple organellar group II intron targets. Moreover, they seem to function in the splicing of group II introns together with a number of additional nuclear-encoded splicing factors, possibly acting as an organellar proto-spliceosome. Together, this makes them interesting models for the early evolution of nuclear spliceosomal splicing. In this review, we summarize recent advances in our understanding of the role of plant maturases and their accessory factors in plants. This article is part of a Special Issue entitled: Chloroplast Biogenesis.


Subject(s)
Endoribonucleases/physiology , Evolution, Molecular , Nucleotidyltransferases/physiology , Spliceosomes/physiology , Cell Nucleus/enzymology , Chloroplasts/enzymology , Introns , Mitochondria/enzymology , RNA Splicing
SELECTION OF CITATIONS
SEARCH DETAIL