Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Immunity ; 57(8): 1812-1827.e7, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38955184

ABSTRACT

An important property of the host innate immune response during microbial infection is its ability to control the expression of antimicrobial effector proteins, but how this occurs post-transcriptionally is not well defined. Here, we describe a critical antibacterial role for the classic antiviral gene 2'-5'-oligoadenylate synthetase 1 (OAS1). Human OAS1 and its mouse ortholog, Oas1b, are induced by interferon-γ and protect against cytosolic bacterial pathogens such as Francisella novicida and Listeria monocytogenes in vitro and in vivo. Proteomic and transcriptomic analysis showed reduced IRF1 protein expression in OAS1-deficient cells. Mechanistically, OAS1 binds and localizes IRF1 mRNA to the rough endoplasmic reticulum (ER)-Golgi endomembranes, licensing effective translation of IRF1 mRNA without affecting its transcription or decay. OAS1-dependent translation of IRF1 leads to the enhanced expression of antibacterial effectors, such as GBPs, which restrict intracellular bacteria. These findings uncover a noncanonical function of OAS1 in antibacterial innate immunity.


Subject(s)
2',5'-Oligoadenylate Synthetase , Immunity, Innate , Interferon Regulatory Factor-1 , 2',5'-Oligoadenylate Synthetase/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Animals , Humans , Mice , Protein Biosynthesis/immunology , Listeria monocytogenes/immunology , Mice, Knockout , Mice, Inbred C57BL , Listeriosis/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology
2.
Immunity ; 57(3): 446-461.e7, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38423012

ABSTRACT

In response to viral infection, how cells balance translational shutdown to limit viral replication and the induction of antiviral components like interferons (IFNs) is not well understood. Moreover, how distinct isoforms of IFN-induced oligoadenylate synthetase 1 (OAS1) contribute to this antiviral response also requires further elucidation. Here, we show that human, but not mouse, OAS1 inhibits SARS-CoV-2 replication through its canonical enzyme activity via RNase L. In contrast, both mouse and human OAS1 protect against West Nile virus infection by a mechanism distinct from canonical RNase L activation. OAS1 binds AU-rich elements (AREs) of specific mRNAs, including IFNß. This binding leads to the sequestration of IFNß mRNA to the endomembrane regions, resulting in prolonged half-life and continued translation. Thus, OAS1 is an ARE-binding protein with two mechanisms of antiviral activity: driving inhibition of translation but also a broader, non-canonical function of protecting IFN expression from translational shutdown.


Subject(s)
2',5'-Oligoadenylate Synthetase , Interferons , Oligoribonucleotides , Virus Diseases , West Nile Fever , Animals , Humans , Mice , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Adenine Nucleotides , Antiviral Agents/pharmacology , West Nile Fever/genetics , West Nile Fever/metabolism , West Nile virus/metabolism , West Nile virus/pathogenicity
3.
Immunity ; 51(3): 548-560.e4, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471106

ABSTRACT

Immunotherapy can reinvigorate dormant responses to cancer, but response rates remain low. Oncolytic viruses, which replicate in cancer cells, induce tumor lysis and immune priming, but their immune consequences are unclear. We profiled the infiltrate of aggressive melanomas induced by oncolytic Vaccinia virus using RNA sequencing and found substantial remodeling of the tumor microenvironment, dominated by effector T cell influx. However, responses to oncolytic viruses were incomplete due to metabolic insufficiencies induced by the tumor microenvironment. We identified the adipokine leptin as a potent metabolic reprogramming agent that supported antitumor responses. Leptin metabolically reprogrammed T cells in vitro, and melanoma cells expressing leptin were immunologically controlled in mice. Engineering oncolytic viruses to express leptin in tumor cells induced complete responses in tumor-bearing mice and supported memory development in the tumor infiltrate. Thus, leptin can provide metabolic support to tumor immunity, and oncolytic viruses represent a platform to deliver metabolic therapy.


Subject(s)
Leptin/immunology , Melanoma/immunology , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Immunotherapy/methods , Male , Mice , Mice, Inbred C57BL , Tumor Microenvironment/immunology , Vaccinia virus/immunology
4.
Immunity ; 51(3): 451-464.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471108

ABSTRACT

Type I and III interferons (IFNs) activate similar downstream signaling cascades, but unlike type I IFNs, type III IFNs (IFNλ) do not elicit strong inflammatory responses in vivo. Here, we examined the molecular mechanisms underlying this disparity. Type I and III IFNs displayed kinetic differences in expression of IFN-stimulated genes and proinflammatory responses, with type I IFNs preferentially stimulating expression of the transcription factor IRF1. Type III IFNs failed to induce IRF1 expression because of low IFNλ receptor abundance and insufficient STAT1 activation on epithelial cells and thus did not activate the IRF1 proinflammatory gene program. Rather, IFNλ stimulation preferentially induced factors implicated in tissue repair. Our findings suggest that IFN receptor compartmentalization and abundance confer a spatiotemporal division of labor where type III IFNs control viral spread at the site of the infection while restricting tissue damage; the transient induction of inflammatory responses by type I IFNs recruits immune effectors to promote protective immunity.


Subject(s)
Interferon Regulatory Factor-1/immunology , Interferon Type I/immunology , Interferons/immunology , Animals , Cell Line , Epithelial Cells/immunology , Humans , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , STAT1 Transcription Factor/immunology , Interferon Lambda
5.
Immunity ; 50(1): 51-63.e5, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30635239

ABSTRACT

Interferon-inducible human oligoadenylate synthetase-like (OASL) and its mouse ortholog, Oasl2, enhance RNA-sensor RIG-I-mediated type I interferon (IFN) induction and inhibit RNA virus replication. Here, we show that OASL and Oasl2 have the opposite effect in the context of DNA virus infection. In Oasl2-/- mice and OASL-deficient human cells, DNA viruses such as vaccinia, herpes simplex, and adenovirus induced increased IFN production, which resulted in reduced virus replication and pathology. Correspondingly, ectopic expression of OASL in human cells inhibited IFN induction through the cGAS-STING DNA-sensing pathway. cGAS was necessary for the reduced DNA virus replication observed in OASL-deficient cells. OASL directly and specifically bound to cGAS independently of double-stranded DNA, resulting in a non-competitive inhibition of the second messenger cyclic GMP-AMP production. Our findings define distinct mechanisms by which OASL differentially regulates host IFN responses during RNA and DNA virus infection and identify OASL as a negative-feedback regulator of cGAS.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , DNA Virus Infections/immunology , DNA Viruses/physiology , RNA Virus Infections/immunology , RNA Viruses/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Cyclic AMP/metabolism , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/metabolism , RNA, Small Interfering/genetics , Signal Transduction , THP-1 Cells , Virus Replication
6.
Immunity ; 49(3): 413-426.e5, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30170814

ABSTRACT

Inflammasome-activated caspase-1 cleaves gasdermin D to unmask its pore-forming activity, the predominant consequence of which is pyroptosis. Here, we report an additional biological role for gasdermin D in limiting cytosolic DNA surveillance. Cytosolic DNA is sensed by Aim2 and cyclic GMP-AMP synthase (cGAS) leading to inflammasome and type I interferon responses, respectively. We found that gasdermin D activated by the Aim2 inflammasome suppressed cGAS-driven type I interferon response to cytosolic DNA and Francisella novicida in macrophages. Similarly, interferon-ß (IFN-ß) response to F. novicida infection was elevated in gasdermin D-deficient mice. Gasdermin D-mediated negative regulation of IFN-ß occurred in a pyroptosis-, interleukin-1 (IL-1)-, and IL-18-independent manner. Mechanistically, gasdermin D depleted intracellular potassium (K+) via membrane pores, and this K+ efflux was necessary and sufficient to inhibit cGAS-dependent IFN-ß response. Thus, our findings have uncovered an additional interferon regulatory module involving gasdermin D and K+ efflux.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Francisella/physiology , Gram-Negative Bacterial Infections/immunology , Inflammasomes/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , DNA Damage , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Interferon Type I/metabolism , Interleukin-1/metabolism , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Phosphate-Binding Proteins , Potassium/metabolism , RNA, Small Interfering/genetics
7.
Mol Cell ; 76(1): 11-26.e7, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31400850

ABSTRACT

Alternative lengthening of telomeres (ALT) is a homology-directed repair (HDR) mechanism of telomere elongation that controls proliferation in aggressive cancers. We show that the disruption of RAD51-associated protein 1 (RAD51AP1) in ALT+ cancer cells leads to generational telomere shortening. This is due to RAD51AP1's involvement in RAD51-dependent homologous recombination (HR) and RAD52-POLD3-dependent break induced DNA synthesis. RAD51AP1 KO ALT+ cells exhibit telomere dysfunction and cytosolic telomeric DNA fragments that are sensed by cGAS. Intriguingly, they activate ULK1-ATG7-dependent autophagy as a survival mechanism to mitigate DNA damage and apoptosis. Importantly, RAD51AP1 protein levels are elevated in ALT+ cells due to MMS21 associated SUMOylation. Mutation of a single SUMO-targeted lysine residue perturbs telomere dynamics. These findings indicate that RAD51AP1 is an essential mediator of the ALT mechanism and is co-opted by post-translational mechanisms to maintain telomere length and ensure proliferation of ALT+ cancer cells.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Telomere Homeostasis , Telomere/metabolism , Autophagy , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Proliferation , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Homologous Recombination , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ligases/genetics , Ligases/metabolism , Lysine , Neoplasms/genetics , Neoplasms/pathology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Stability , RNA-Binding Proteins/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Signal Transduction , Sumoylation , Telomere/genetics , Telomere/pathology
8.
PLoS Pathog ; 20(2): e1011840, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38315735

ABSTRACT

Human metapneumovirus (HMPV) is an important cause of acute lower respiratory infection in children and adults worldwide. There are four genetic subgroups of HMPV and both neutralizing antibodies and T cells contribute to protection. However, little is known about mechanisms of pathogenesis and most published work is based on a few extensively passaged, laboratory-adapted strains of HMPV. In this study, we isolated and characterized a panel of low passage HMPV clinical isolates representing all four genetic subgroups. The clinical isolates exhibited lower levels of in vitro replication compared to a lab-adapted strain. We compared disease phenotypes using a well-established mouse model. Several virulent isolates caused severe weight loss, lung pathology, airway dysfunction, and fatal disease in mice, which was confirmed in three inbred mouse strains. Disease severity did not correlate with lung viral titer, as virulent strains exhibited restricted replication in the lower airway. Virulent HMPV isolates were associated with markedly increased proinflammatory cytokine production and neutrophil influx; however, depletion of neutrophils or genetic ablation of inflammasome components did not reverse disease. Virulent clinical isolates induced markedly increased type I and type III interferon (IFN) secretion in vitro and in vivo. STAT1/2-deficient mice lacking both type I and type III IFN signaling showed reduced disease severity and increased lung viral replication. Inhibition of type I IFN signaling using a blocking antibody or genetic ablation of the type I IFN receptor reduced pathology with minimal effect on viral replication. Conversely, blockade of type III IFN signaling with a neutralizing antibody or genetic ablation of the IFN-lambda receptor had no effect on pathogenesis but restored viral replication. Collectively, these results demonstrate distinct roles for type I and type III IFN in HMPV pathogenesis and immunity.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Respiratory Tract Infections , Child , Animals , Mice , Humans , Interferon Lambda , Lung , Respiratory Tract Infections/pathology , Interferons
9.
Immunity ; 40(6): 936-48, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24931123

ABSTRACT

Virus infection is sensed in the cytoplasm by retinoic acid-inducible gene I (RIG-I, also known as DDX58), which requires RNA and polyubiquitin binding to induce type I interferon (IFN) and activate cellular innate immunity. We show that the human IFN-inducible oligoadenylate synthetases-like (OASL) protein has antiviral activity and mediates RIG-I activation by mimicking polyubiquitin. Loss of OASL expression reduced RIG-I signaling and enhanced virus replication in human cells. Conversely, OASL expression suppressed replication of a number of viruses in a RIG-I-dependent manner and enhanced RIG-I-mediated IFN induction. OASL interacted and colocalized with RIG-I, and through its C-terminal ubiquitin-like domain specifically enhanced RIG-I signaling. Bone-marrow-derived macrophages from mice deficient for Oasl2 showed that among the two mouse orthologs of human OASL, Oasl2 is functionally similar to human OASL. Our findings show a mechanism by which human OASL contributes to host antiviral responses by enhancing RIG-I activation.


Subject(s)
2',5'-Oligoadenylate Synthetase/immunology , DEAD-box RNA Helicases/immunology , DNA Virus Infections/immunology , Interferon Type I/immunology , RNA Virus Infections/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , DEAD Box Protein 58 , HCT116 Cells , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyubiquitin , Protein Binding/immunology , RNA Interference , RNA, Small Interfering , Receptors, Immunologic , Signal Transduction/immunology , Virus Replication/immunology
12.
J Theor Biol ; 462: 148-157, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30395807

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) has recently been identified as the primary protein that detects cytosolic double stranded DNA to invoke a type I interferon response. The cGAS pathway is vital in the recognition of DNA encoded viruses as well as self-DNA leaked from the nucleus of damaged cells. Currently, the dynamics regulating the cGAS pathway are poorly understood; limiting our knowledge of how DNA-induced immune responses are regulated. Using systems biology approaches, we formulated a mathematical model to describe the dynamics of this pathway and examine the resulting system-level emergent properties. Unknown model parameters were fit to data compiled from literature using a Parallel Tempering Markov Chain Monte Carlo (PT-MCMC) approach, resulting in an ensemble of parameterized models. A local sensitivity analysis demonstrated that parameter sensitivity trends across model ensembles were independent of the select parameterization. An in-silico knock-down of TREX1 found that the interferon response is highly robust, showing that complete inhibition is necessary to induce chemical conditions consistent with chronic inflammation. Lastly, we demonstrate that the model recapitulates interferon expression data resulting from small molecule inhibition of cGAS. Overall, the importance of this model is exhibited in its capacity to identify sensitive components of the cGAS pathway, generate testable hypotheses, and confirm experimental observations.


Subject(s)
DNA/immunology , Exodeoxyribonucleases/metabolism , Models, Theoretical , Nucleotidyltransferases/metabolism , Phosphoproteins/metabolism , Animals , DNA, Viral/immunology , Feedback , Humans , Inflammation , Interferon Type I/metabolism , Markov Chains , Monte Carlo Method , Systems Biology/methods
13.
Proc Natl Acad Sci U S A ; 113(6): 1642-7, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26729873

ABSTRACT

Clinical observations link respiratory virus infection and Pseudomonas aeruginosa colonization in chronic lung disease, including cystic fibrosis (CF) and chronic obstructive pulmonary disease. The development of P. aeruginosa into highly antibiotic-resistant biofilm communities promotes airway colonization and accounts for disease progression in patients. Although clinical studies show a strong correlation between CF patients' acquisition of chronic P. aeruginosa infections and respiratory virus infection, little is known about the mechanism by which chronic P. aeruginosa infections are initiated in the host. Using a coculture model to study the formation of bacterial biofilm formation associated with the airway epithelium, we show that respiratory viral infections and the induction of antiviral interferons promote robust secondary P. aeruginosa biofilm formation. We report that the induction of antiviral IFN signaling in response to respiratory syncytial virus (RSV) infection induces bacterial biofilm formation through a mechanism of dysregulated iron homeostasis of the airway epithelium. Moreover, increased apical release of the host iron-binding protein transferrin during RSV infection promotes P. aeruginosa biofilm development in vitro and in vivo. Thus, nutritional immunity pathways that are disrupted during respiratory viral infection create an environment that favors secondary bacterial infection and may provide previously unidentified targets to combat bacterial biofilm formation.


Subject(s)
Biofilms/growth & development , Immunity , Nutritional Physiological Phenomena , Pseudomonas aeruginosa/physiology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/physiology , Animals , Antiviral Agents/pharmacology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/virology , Homeostasis/drug effects , Humans , Interferon-beta/pharmacology , Iron/pharmacology , Mice , Microbial Interactions/drug effects , Models, Biological , Pseudomonas aeruginosa/drug effects , Respiratory Syncytial Viruses/drug effects , Signal Transduction/drug effects , Transferrin/metabolism
14.
J Immunol ; 196(9): 3877-86, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27016603

ABSTRACT

Moloney leukemia virus 10, homolog (MOV10) is an IFN-inducible RNA helicase, associated with small RNA-induced silencing. In this article, we report that MOV10 exhibits antiviral activity, independent of its helicase function, against a number of positive- and negative-strand RNA viruses by enhancing type I IFN induction. Using a number of genome-edited knockout human cells, we show that IFN regulatory factor 3-mediated IFN induction and downstream IFN signaling through IFN receptor was necessary to inhibit virus replication by MOV10. MOV10 enhanced IFN regulatory factor 3-mediated transcription of IFN. However, this IFN induction by MOV10 was unique and independent of the known retinoic acid-inducible gene I/mitochondrial antiviral-signaling protein-mediated RNA-sensing pathway. Upon virus infection, MOV10 specifically required inhibitor of κB kinase ε, not TANK-binding kinase 1, for its antiviral activity. The important role of MOV10 in mediating antiviral signaling was further supported by the finding that viral proteases from picornavirus family specifically targeted MOV10 as a possible innate immune evasion mechanism. These results establish MOV10, an evolutionary conserved protein involved in RNA silencing, as an antiviral gene against RNA viruses that uses an retinoic acid-inducible gene I-like receptor-independent pathway to enhance IFN response.


Subject(s)
Cardiovirus Infections/immunology , Encephalomyocarditis virus/immunology , RNA Helicases/metabolism , Rhabdoviridae Infections/immunology , Vesicular stomatitis Indiana virus/immunology , Adaptor Proteins, Signal Transducing/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Immune Evasion , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , RNA Helicases/genetics , RNA Interference , RNA, Viral/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction
15.
PLoS Pathog ; 11(9): e1005150, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26372645

ABSTRACT

Interferon stimulated genes (ISGs) target viruses at various stages of their infectious life cycles, including at the earliest stage of viral entry. Here we identify ArfGAP with dual pleckstrin homology (PH) domains 2 (ADAP2) as a gene upregulated by type I IFN treatment in a STAT1-dependent manner. ADAP2 functions as a GTPase-activating protein (GAP) for Arf6 and binds to phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P3) and PI(3,4)P2. We show that overexpression of ADAP2 suppresses dengue virus (DENV) and vesicular stomatitis virus (VSV) infection in an Arf6 GAP activity-dependent manner, while exerting no effect on coxsackievirus B (CVB) or Sendai virus (SeV) replication. We further show that ADAP2 expression induces macropinocytosis and that ADAP2 strongly associates with actin-enriched membrane ruffles and with Rab8a- and LAMP1-, but not EEA1- or Rab7-, positive vesicles. Utilizing two techniques--light-sensitive neutral red (NR)-containing DENV and fluorescence assays for virus internalization--we show that ADAP2 primarily restricts DENV infection at the stage of virion entry and/or intracellular trafficking and that incoming DENV and VSV particles associate with ADAP2 during their entry. Taken together, this study identifies ADAP2 as an ISG that exerts antiviral effects against RNA viruses by altering Arf6-mediated trafficking to disrupt viral entry.


Subject(s)
ADP-Ribosylation Factors/agonists , Dengue Virus/physiology , GTPase-Activating Proteins/agonists , Interferon-beta/metabolism , Pinocytosis , Vesiculovirus/physiology , Virus Internalization , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Cell Line , Chlorocebus aethiops , Dengue Virus/immunology , Enterovirus B, Human/immunology , Enterovirus B, Human/physiology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Mutation , Phosphatidylinositol Phosphates/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Sendai virus/immunology , Sendai virus/physiology , Vero Cells , Vesiculovirus/immunology , Virion/immunology , Virion/physiology , Virus Replication
16.
Nucleic Acids Res ; 43(10): 5236-48, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25925578

ABSTRACT

The oligoadenylate synthetase (OAS) enzymes are cytoplasmic dsRNA sensors belonging to the antiviral innate immune system. Upon binding to viral dsRNA, the OAS enzymes synthesize 2'-5' linked oligoadenylates (2-5As) that initiate an RNA decay pathway to impair viral replication. The human OAS-like (OASL) protein, however, does not harbor the catalytic activity required for synthesizing 2-5As and differs from the other human OAS family members by having two C-terminal ubiquitin-like domains. In spite of its lack of enzymatic activity, human OASL possesses antiviral activity. It was recently demonstrated that the ubiquitin-like domains of OASL could substitute for K63-linked poly-ubiquitin and interact with the CARDs of RIG-I and thereby enhance RIG-I signaling. However, the role of the OAS-like domain of OASL remains unclear. Here we present the crystal structure of the OAS-like domain, which shows a striking similarity with activated OAS1. Furthermore, the structure of the OAS-like domain shows that OASL has a dsRNA binding groove. We demonstrate that the OAS-like domain can bind dsRNA and that mutating key residues in the dsRNA binding site is detrimental to the RIG-I signaling enhancement. Hence, binding to dsRNA is an important feature of OASL that is required for enhancing RIG-I signaling.


Subject(s)
2',5'-Oligoadenylate Synthetase/chemistry , DEAD-box RNA Helicases/metabolism , RNA, Double-Stranded/chemistry , RNA-Binding Proteins/chemistry , 2',5'-Oligoadenylate Synthetase/metabolism , DEAD Box Protein 58 , HEK293 Cells , Humans , Models, Molecular , Protein Multimerization , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Receptors, Immunologic , Signal Transduction
17.
Carcinogenesis ; 37(5): 522-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26992898

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a devastating disease for which new treatments, such as immunotherapy are needed. Synthetic double-stranded RNAs, which activate toll-like receptor 3 (TLR3), have been used as potent adjuvants in cancer immunotherapy by triggering a proapoptotic response in cancer cells. A better understanding of the mechanism of TLR3-mediated apoptosis and its potential involvement in controlling tumor metastasis could lead to improvements in current treatment. Using paired, autologous primary and metastatic HNSCC cells we previously showed that metastatic, but not primary tumor-derived cells, were unable to activate prosurvival NF-κB in response to p(I):p(C) resulting in an enhanced apoptotic response. Here, we show that transcriptional downregulation of receptor-interacting serine/threonine-protein kinase 1 (RIPK1) in metastatic HNSCC cells causes a loss of TLR3-mediated NF-κB signaling, resulting in enhanced apoptosis. Loss of RIPK1 strongly correlates with metastatic disease in a cohort of HNSCC patients. This downregulation of RIPK1 is possibly mediated by enhanced methylation of the RIPK1 promoter in tumor cells and enhances protumorigenic properties such as cell migration. The results described here establish a novel mechanism of TLR3-mediated apoptosis in metastatic cells and may create new opportunities for using double stranded RNA to target metastatic tumor cells.


Subject(s)
Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Apoptosis/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA Methylation , Down-Regulation , Head and Neck Neoplasms/metabolism , Humans , Immunity, Innate/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA, Double-Stranded/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Toll-Like Receptor 3/metabolism
18.
J Virol ; 89(19): 10115-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26178980

ABSTRACT

2'-5'-Oligoadenylate synthetase-like protein (OASL) is an interferon-inducible antiviral protein. Here we describe differential inhibitory activities of human OASL and the two mouse OASL homologs against respiratory syncytial virus (RSV) replication. Interestingly, nonstructural protein 1 (NS1) of RSV promoted proteasome-dependent degradation of specific OASL isoforms. We conclude that OASL acts as a cellular antiviral protein and that RSV NS1 suppresses this function to evade cellular innate immunity and allow virus growth.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Respiratory Syncytial Viruses/physiology , Viral Nonstructural Proteins/physiology , Virus Replication/physiology , 2',5'-Oligoadenylate Synthetase/immunology , Animals , HEK293 Cells , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Cellular , Mice , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Viral Nonstructural Proteins/genetics , Virus Replication/immunology
19.
J Immunol ; 192(12): 5933-42, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24799566

ABSTRACT

Polyomaviruses encode a large T Ag (LT), a multifunctional protein essential for the regulation of both viral and host cell gene expression and productive viral infection. Previously, we have shown that stable expression of LT protein results in upregulation of genes involved in the IFN induction and signaling pathway. In this study, we focus on the cellular signaling mechanism that leads to the induction of IFN responses by LT. Our results show that ectopic expression of SV40 LT results in the induction of IFN-stimulated genes (ISGs) in human fibroblasts and confers an antiviral state. We describe a LT-initiated DNA damage response (DDR) that activates IFN regulatory factor 1, causing IFN-ß production and consequent ISG expression in human cells. This IFN-ß and ISG induction is dependent on ataxia-telangiectasia mutated and Rad3-related (ATR) kinase, but independent of ATM. ATR kinase inhibition using a selective kinase inhibitor (ETP-46464) caused a decrease in IFN regulatory factor 1 stabilization and ISG expression. Furthermore, expression of a mutant LT that does not induce DDR also does not induce IFN-ß and ISGs. These results show that, in the absence of viral infection, LT-initiated activation of ATR-dependent DDR is sufficient for the induction of an IFN-ß-mediated innate immune response in human cells. Thus, we have uncovered a novel and critical role for ATR as a mediator of antiviral responses utilizing LT.


Subject(s)
Antigens, Polyomavirus Transforming/immunology , DNA Damage/immunology , Interferon Regulatory Factor-1/immunology , Interferon-beta/immunology , Simian virus 40/immunology , Antigens, Polyomavirus Transforming/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/immunology , DNA Damage/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon-beta/genetics , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , Simian virus 40/genetics
20.
J Immunol ; 193(7): 3436-45, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172485

ABSTRACT

Type I IFNs play central roles in innate immunity; however, overproduction of IFN can lead to immunopathology. In this study, we demonstrate that adenosine deaminase acting on RNA 1 (ADAR1), an RNA-editing enzyme induced by IFN, is essential for cells to avoid inappropriate sensing of cytosolic RNA in an inducible knockout cell model-the primary mouse embryo fibroblast derived from ADAR1 lox/lox and Cre-ER mice as well as in HEK293 cells. ADAR1 suppresses viral and cellular RNA detection by retinoic acid-inducible gene I (RIG-I) through its RNA binding rather than its RNA editing activity. dsRNA binds to both ADAR1 and RIG-I, but ADAR1 reduces RIG-I RNA binding. In the absence of ADAR1, cellular RNA stimulates type I IFN production without viral infection or exogenous RNA stimulation. Moreover, we showed in the ADAR1-inducible knockout mice that ADAR1 gene disruption results in high-level IFN production in neuronal tissues-the hallmark of Aicardi-Goutières syndrome, a heritable autoimmune disease recently found to be associated with ADAR1 gene mutations. In summary, this study found that ADAR1 limits cytosolic RNA sensing by RIG-I through its RNA binding activity; therefore, ADAR1 suppresses type I IFN production stimulated by viral and cellular RNAs. These results explain why loss of ADARA1 causes IFN induction and also indicates a mechanism for the involvement of ADAR1 in autoimmune diseases such as Aicardi-Goutières syndrome.


Subject(s)
Adenosine Deaminase/immunology , DEAD-box RNA Helicases/immunology , DEAD-box RNA Helicases/metabolism , Interferon Type I/immunology , RNA, Viral/immunology , Sendai virus/immunology , Adenosine Deaminase/genetics , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon Type I/genetics , Mice , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Nervous System Malformations/pathology , RNA, Viral/genetics , RNA-Binding Proteins , Receptors, Immunologic , Sendai virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL