Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Nat Med ; 27(4): 659-667, 2021 04.
Article in English | MEDLINE | ID: mdl-33633408

ABSTRACT

To identify circulating proteins influencing Coronavirus Disease 2019 (COVID-19) susceptibility and severity, we undertook a two-sample Mendelian randomization (MR) study, rapidly scanning hundreds of circulating proteins while reducing bias due to reverse causation and confounding. In up to 14,134 cases and 1.2 million controls, we found that an s.d. increase in OAS1 levels was associated with reduced COVID-19 death or ventilation (odds ratio (OR) = 0.54, P = 7 × 10-8), hospitalization (OR = 0.61, P = 8 × 10-8) and susceptibility (OR = 0.78, P = 8 × 10-6). Measuring OAS1 levels in 504 individuals, we found that higher plasma OAS1 levels in a non-infectious state were associated with reduced COVID-19 susceptibility and severity. Further analyses suggested that a Neanderthal isoform of OAS1 in individuals of European ancestry affords this protection. Thus, evidence from MR and a case-control study support a protective role for OAS1 in COVID-19 adverse outcomes. Available pharmacological agents that increase OAS1 levels could be prioritized for drug development.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , COVID-19/etiology , Genetic Predisposition to Disease , SARS-CoV-2 , 2',5'-Oligoadenylate Synthetase/genetics , Aged , Aged, 80 and over , Animals , COVID-19/genetics , Case-Control Studies , Female , Humans , Interleukin-10 Receptor beta Subunit/genetics , Male , Mendelian Randomization Analysis , Middle Aged , Neanderthals , Protein Isoforms/physiology , Quantitative Trait Loci , Severity of Illness Index , White People
2.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462564

ABSTRACT

In mice, resistance to central nervous system (CNS) disease induced by members of the genus Flavivirus is conferred by an allele of the 2'-5' oligoadenylate synthetase 1b gene that encodes the inactive full-length protein (Oas1b-FL). The susceptibility allele encodes a C-terminally truncated protein (Oas1b-tr). We show that the efficiency of neuron infection in the brains of resistant and susceptible mice is similar after an intracranial inoculation of two flaviviruses, but amplification of viral proteins and double-stranded RNA (dsRNA) is inhibited in infected neurons in resistant mouse brains at later times. Active OAS proteins detect cytoplasmic dsRNA and synthesize short 2'-5'-linked oligoadenylates (2'-5'A) that interact with the latent endonuclease RNase L, causing it to dimerize and cleave single-stranded RNAs. To evaluate the contribution of RNase L to the resistance phenotype in vivo, we created a line of resistant RNase L-/- mice. Evidence of RNase L activation in infected RNase L+/+ mice was indicated by higher levels of viral RNA in the brains of infected RNase L-/- mice. Activation of type I interferon (IFN) signaling was detected in both resistant and susceptible brains, but Oas1a and Oas1b mRNA levels were lower in RNase L+/+ mice of both types, suggesting that activated RNase L also has a proflaviviral effect. Inhibition of virus replication was robust in resistant RNase L-/- mice, indicating that activated RNase L is not a critical factor in mediating this phenotype.IMPORTANCE The mouse genome encodes a family of Oas proteins that synthesize 2'-5'A in response to dsRNA. 2'-5'A activates the endonuclease RNase L to cleave single-stranded viral and cellular RNAs. The inactive, full-length Oas1b protein confers flavivirus-specific disease resistance. Although similar numbers of neurons were infected in resistant and susceptible brains after an intracranial virus infection, viral components amplified only in susceptible brains at later times. A line of resistant RNase L-/- mice was used to evaluate the contribution of RNase L to the resistance phenotype in vivo Activation of RNase L antiviral activity by flavivirus infection was indicated by increased viral RNA levels in the brains of RNase L-/- mice. Oas1a and Oas1b mRNA levels were higher in infected RNase L-/- mice, indicating that activated RNase L also have a proflaviviral affect. However, the resistance phenotype was equally robust in RNase L-/- and RNase L+/+ mice.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Endoribonucleases/metabolism , Flavivirus Infections/metabolism , 2',5'-Oligoadenylate Synthetase/physiology , Adenine Nucleotides/genetics , Adenine Nucleotides/metabolism , Animals , Cell Line , Endoribonucleases/genetics , Endoribonucleases/physiology , Flavivirus/metabolism , Flavivirus Infections/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Phenotype , RNA, Viral/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism , Virus Replication/drug effects
3.
Vet Immunol Immunopathol ; 209: 22-30, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30885302

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) has previously been shown to increase porcine 2'-5'-oligoadenylate synthase (OAS) 1a expression, but the specific role of porcine OAS1b (pOAS1b) in PRRSV replication remains unknown. In this study, we conducted sequence analysis of the porcine OAS1b gene and studied the effects of its overexpression or silencing on PRRSV replication. OAS1b, localized mainly in the cytoplasm, was found to contain conserved protein domains, such as the P-Loop and D-Box, indicating that its nucleotidyl transferase activity was complete and the antiviral effect depended on ribonuclease L (RNase L). OAS1b overexpression inhibited PRRSV replication, whereas small-interfering-RNA silencing of OAS1b resulted in increased virus titers. Additionally, OAS1b promoted expression of interferons as well as interferon-ß promoter activity. These results lay the theoretical foundation for the development of new anti-PRRSV strategies.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/physiology , Swine Diseases/virology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Cells, Cultured , Cloning, Molecular , Gene Silencing , Humans , RNA, Messenger/metabolism , Sequence Analysis, Protein , Species Specificity , Swine , Tissue Distribution , Transcription Factors/metabolism , Virus Replication/genetics
5.
Article in English | MEDLINE | ID: mdl-28580319

ABSTRACT

The type I IFN response quickly became associated with its role in the innate immune response to viral infection. The past few years have seen the significance of IFNs expand in breadth to include non-viral pathogens. Previous work has identified that following viral infection, type I IFN signaling induces the production of the 2'-5'-oligoadenylate synthetase (OAS) family, which include OAS1, OAS2, OAS3, and OAS-like (OASL) protein. OASL was identified to be strongly induced following viral infection through engaging the RNA sensor RIG-I and increasing signaling through this pathway to enhance the anti-viral type I IFN response. Surprisingly, infection with viral dsDNA revealed an IFN inhibitory role and therefore pro-viral function of OASL through the inhibition of the cGAS cytosolic DNA sensing mechanism. Intracellular bacteria are able to activate the cytosolic DNA sensing pathway, however the role of OASL during bacterial infection is largely unknown. Vacuolar pathogenic microbes such as mycobacteria induce OASL early post infection, where it functions in a prosurvival fashion by inhibiting autophagic mechanisms and antimicrobial peptide expression. This suggests an underestimated role of OASL in the innate immune response to infection with a variety of pathogens and points to OASL-associated modulation of the type I IFN response. OASL may therefore play a critical role in defining the outcome of infection. We provide a brief update on the recent developments of the OAS family of proteins in response to DNA and RNA virus infections, as well as discuss evidence of Oasl expression in response to a number of cytosolic and vacuolar replicating bacterial pathogens.


Subject(s)
2',5'-Oligoadenylate Synthetase/immunology , Bacteria/immunology , Cytoplasm/immunology , Host-Pathogen Interactions/immunology , Interferon Type I/immunology , 2',5'-Oligoadenylate Synthetase/classification , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/physiology , Autophagy/physiology , Bacteria/growth & development , Bacteria/pathogenicity , Cytoplasm/genetics , Cytoplasm/microbiology , Cytosol/immunology , Cytosol/microbiology , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate , Interferon Type I/genetics , Mycobacterium/immunology , Mycobacterium/pathogenicity , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Signal Transduction/immunology , Vacuoles/microbiology , Virus Diseases/immunology
6.
Neurosci Lett ; 628: 78-84, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27297771

ABSTRACT

Type I Interferon (IFN-I) is critical for antiviral and antitumor defense. Additionally, IFN-I has been used for treating multiple sclerosis (MS), a chronic autoimmune disease of the central nervous system (CNS). Recently, we reported that 2'-5' oligoadenylate synthetase-like 1 (OASL1) negatively regulates IFN-I production upon viral infection and tumor challenge. Therefore, OASL1 deficient (Oasl1(-)(/)(-)) mice are resistant to viral infections and tumor challenge. In this study, we examined whether OASL1 plays a negative role in the development of autoimmune MS by using Oasl1(-)(/)(-) mice and a murine MS model, myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). Oasl1(-)(/)(-) mice showed enhanced resistance to EAE development compared to wild-type (WT) mice. Additionally, EAE-induced Oasl1(-)(/)(-) mice showed fewer infiltrated immune cells such as T cells and macrophages in the CNS and less CNS inflammation, compared to WT mice. Collectively, these results indicate that OASL1 deficiency suppresses the development of MS-like autoimmunity and suggest that negative regulators of IFN-I could be good therapeutic targets for treating MS in humans.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Cerebellum/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Spinal Cord/pathology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Blood-Brain Barrier/metabolism , Cerebellum/immunology , Cerebellum/metabolism , Disease Models, Animal , Encephalitis/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Spinal Cord/immunology , Spinal Cord/metabolism , T-Lymphocytes/metabolism
7.
Mediators Inflamm ; 2015: 716315, 2015.
Article in English | MEDLINE | ID: mdl-26146465

ABSTRACT

It has been reported that IFN-λs inhibit HCV replication in vitro. But the mechanisms of how IL-28A conducts antiviral activity and the functions of IL-28A-induced ISGs (IFN-stimulated genes) are not fully understood. In this study, we found that IL-28A has the antiviral effect on HCV life cycle including viral replication, assembly, and release. IL-28A and IFN-α synergistically inhibit virus replication. EPSTI1 (epithelial-stromal interaction 1), one of IL-28A-induced ISGs, plays a vital role in IL-28A-mediated antiviral activity. Furthermore, forced expression of EPSTI1 effectively inhibits HCV replication in the absence of interferon treatment, and knockdown of EPSTI1 contributes to viral enhancement. EPSTI1 can activate PKR promoter and induce several PKR-dependent genes, including IFN-ß, IFIT1, OAS1, and RNase L, which is responsible for EPSTI1-mediated antiviral activity.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Interleukins/pharmacology , Neoplasm Proteins/physiology , 2',5'-Oligoadenylate Synthetase/physiology , Cells, Cultured , Hepacivirus/physiology , Humans , Interferon-alpha/pharmacology , Promoter Regions, Genetic , Virus Assembly/drug effects , Virus Replication/drug effects , eIF-2 Kinase/genetics , eIF-2 Kinase/physiology
8.
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
9.
J Hepatol ; 59(6): 1331-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23933585

ABSTRACT

Infections with the hepatitis C virus (HCV) are a major cause of chronic liver disease. While the acute phase of infection is mostly asymptomatic, this virus has the high propensity to establish persistence and in the course of one to several decades liver disease can develop. HCV is a paradigm for the complex interplay between the interferon (IFN) system and viral countermeasures. The virus induces an IFN response within the infected cell and is rather sensitive against the antiviral state triggered by IFNs, yet in most cases HCV persists. Numerous IFN-stimulated genes (ISGs) have been reported to suppress HCV replication, but in only a few cases we begin to understand the molecular mechanisms underlying antiviral activity. It is becoming increasingly clear that blockage of viral replication is mediated by the concerted action of multiple ISGs that target different steps of the HCV replication cycle. This review briefly summarizes the activation of the IFN system by HCV and then focuses on ISGs targeting the HCV replication cycle and their possible mode of action.


Subject(s)
Hepacivirus/physiology , Interferons/physiology , 2',5'-Oligoadenylate Synthetase/physiology , Antigens, Differentiation/physiology , DEAD Box Protein 58 , DEAD-box RNA Helicases/physiology , Hepacivirus/isolation & purification , Humans , Oxidoreductases Acting on CH-CH Group Donors , Proteins/physiology , Receptors, Immunologic , Toll-Like Receptors/physiology , Virus Replication , eIF-2 Kinase/physiology
10.
Vet Immunol Immunopathol ; 153(1-2): 99-106, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23481654

ABSTRACT

In mammals, Toll-like receptor 7 (TLR7) is an important membrane-bound receptor triggered by antiviral compounds and single-stranded RNA. It is implicated in the immune response to viruses such as influenza virus. It was not known whether geese, a natural host for avian influenza viruses, possess a homologue of mammalian TLR7 for recognizing avian influenza virus. In this study, we cloned the full-length of goose TLR7 and partial sequences of its adaptor protein, myeloid differentiation factor 88 (MyD88), some antiviral molecules such as RNA-dependent protein kinase (PKR) and 2',5'-oligoadenylate synthetase (OAS). Goose TLR7 has a protein secondary structure identical to that of mammals, consisting of several leucine-rich domains, a transmembrane domain, and Toll/interleukin-1 receptor domain. To further understand whether the MyD88-dependent pathway of TLR7 is involved in the antiviral innate immune response against highly pathogenic avian influenza virus (HPAIV) infection in geese, we inoculated geese with an H5N1 HPAIV isolated from ducks in 2004. The virus, A/Duck/Guangdong/212/2004, replicated in various tissues resulting in 40% mortality. Quantitative real-time PCR analysis showed upregulation of mRNA transcripts for TLR7, MyD88, PKR and OAS in the lungs of geese at 1, 2 and 3 days post-inoculation. Therefore, the MyD88-dependent pathway of TLR7 was involved in the early stage of antiviral innate immune response in geese during H5N1 HPAIV infection.


Subject(s)
Geese/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/immunology , Myeloid Differentiation Factor 88/physiology , Toll-Like Receptor 7/physiology , 2',5'-Oligoadenylate Synthetase/physiology , Amino Acid Sequence , Animals , Humans , Influenza A Virus, H5N1 Subtype/pathogenicity , Lung/immunology , Mice , Molecular Sequence Data , Toll-Like Receptor 7/chemistry , Toll-Like Receptor 7/genetics , Virus Replication , eIF-2 Kinase/physiology
11.
J Allergy Clin Immunol ; 127(2): 430-438.e1-2, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21208652

ABSTRACT

BACKGROUND: Previous studies have found specificity protein (Sp) 1 transcription factor in the viral replication machinery and postulated that Sp1 was required for viral replication in host cells. OBJECTIVES: We investigated the role of Sp1 in the skin's antiviral responses from the perspective of host defense and its biological relevance in patients with atopic dermatitis and a history of eczema herpeticum (ADEH(+)). METHODS: Small interfering RNA duplexes were used to knock down Sp1 in keratinocytes. The expression of vaccinia virus (VV), herpes simplex virus 1, and other genes were evaluated by real-time PCR, or combined with Western blot and immunohistofluorescence staining. A total of 106 human subjects participated in this study. RESULTS: Both VV and herpes simplex virus 1 replication were enhanced in Sp1 knocked-down keratinocytes. Sp1 gene expression was significantly decreased in ADEH(+) subjects compared with patients with atopic dermatitis without a history of eczema herpeticum and nonatopic subjects (P < .0001) and inversely correlated with VV DNA copy number in human skin explants incubated with VV in vitro (partial correlation r = -0.256; P = .009). Gene profiling revealed that the antiviral genes, double-stranded RNA-dependent protein kinase (PKR) and 2'5'-oligoadenylate synthetase 2 (OAS2), were significantly downregulated in Sp1-silenced keratinocytes. Gene expression of PKR and OAS2 was also significantly decreased in skin biopsies from ADEH(+) subjects compared with patients with atopic dermatitis without a history of eczema herpeticum and nonatopic subjects. IFN-γ augmented the antiviral capacity of Sp1-silenced keratinocytes. CONCLUSION: Specificity protein 1 knockdown enhances viral replication in keratinocytes by downregulating gene expression of PKR and OAS2. Sp1 deficiency in ADEH(+) patients may contribute to their increased propensity to disseminated skin viral infections. IFN-γ augmentation may be a potential treatment for ADEH(+) patients.


Subject(s)
Skin/immunology , Skin/virology , Sp1 Transcription Factor/physiology , 2',5'-Oligoadenylate Synthetase/physiology , Adult , Cells, Cultured , Dermatitis, Atopic/immunology , Dermatitis, Atopic/virology , Eukaryotic Initiation Factor-2/physiology , Female , Gene Silencing , Humans , Interferon-gamma/pharmacology , Kaposi Varicelliform Eruption/immunology , Kaposi Varicelliform Eruption/virology , Keratinocytes/virology , Male , Middle Aged , Sp1 Transcription Factor/genetics , Vaccinia virus/physiology , Virus Replication , eIF-2 Kinase/physiology
12.
Virology ; 399(1): 176-185, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20100623

ABSTRACT

The interferon-inducible 2',5'-oligoadenylate synthetase 1b (Oas1b) protein inhibits West Nile virus (WNV) infection by preventing viral RNA (vRNA) accumulation in infected cells. Serial passage of WNV in Oas1b-expressing mouse cells selected a virus variant with improved growth capacity. Two major amino acid substitutions were identified in this Oas1b-resistant WNV variant: NS3-S365G in the ATPase/helicase domain of NS3 and 2K-V9M in the C-terminal segment of NS4A. To assess their effect on antiviral activity of Oas1b, the NS3 and 2K mutations were engineered into an infectious WNV cDNA clone. The NS3 mutation alters requirement of ATP for ATPase activity and attenuates Oas1b-mediated suppression of vRNA accumulation. However, growth of NS3-mutant virus remains impaired in Oas1b-expressing cells. Only the 2K-V9M mutation efficiently rescued viral growth by promoting vRNA replication. Thus, WNV resistance to Oas1b antiviral action could be attributed to the 2K-V9M substitution with a potential role of NS3-S365G through rescue of vRNA accumulation.


Subject(s)
2',5'-Oligoadenylate Synthetase/pharmacology , 2',5'-Oligoadenylate Synthetase/physiology , DNA Helicases/physiology , West Nile virus/pathogenicity , Amino Acid Substitution/genetics , Animals , DNA Helicases/metabolism , Disease Susceptibility , Gene Expression Regulation, Viral , Mice , Point Mutation/genetics , RNA, Viral/genetics , Virus Replication/physiology , West Nile Fever/drug therapy , West Nile Fever/genetics , West Nile Fever/virology , West Nile virus/genetics , West Nile virus/physiology
13.
J Immunol ; 183(12): 8035-43, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19923450

ABSTRACT

The 2',5'-oligoadenylate synthetase (OAS) and its downstream effector RNase L play important roles in host defense against virus infection. Oas1b, one of the eight Oas1 genes in the mouse genome, has been identified as a murine flavivirus-resistance gene. Four genes, OAS1, OAS2, OAS3, and OAS-like (OASL), have been identified in the human OAS gene family, and 10 isoforms, including OAS1 (p42, p44, p46, p48, and p52), OAS2 (p69 and p71), OAS3 (p100), and OASL (p30 and p59) can be generated by alternative splicing. In this study, we determined the role of the human OAS/RNase L pathway in host defense against dengue virus (DEN) infection and assessed the antiviral potential of each isoform in the human OAS family. DEN replication was reduced by overexpression and enhanced by knockdown of RNase L expression, indicating a protective role for RNase L against DEN replication in human cells. The human OAS1 p42, OAS1 p46, and OAS3 p100, but not the other OAS isoforms, blocked DEN replication via an RNase L-dependent mechanism. Furthermore, the anti-DEN activities of these three OAS isoforms correlated with their ability to trigger RNase L activation in DEN-infected cells. Thus, OAS1 p42/p46 and OAS3 p100 are likely to contribute to host defense against DEN infection and play a role in determining the outcomes of DEN disease severity.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Dengue Virus/immunology , Dengue/enzymology , Dengue/prevention & control , Multigene Family , 2',5'-Oligoadenylate Synthetase/biosynthesis , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Cell Line , Cell Line, Tumor , Cricetinae , Dengue/immunology , Dengue/virology , Endoribonucleases/genetics , Endoribonucleases/metabolism , Endoribonucleases/physiology , Enzyme Activation/immunology , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Virus Activation/immunology
14.
Curr Top Microbiol Immunol ; 321: 85-100, 2008.
Article in English | MEDLINE | ID: mdl-18727488

ABSTRACT

Mouse susceptibility to experimental infections with flaviviruses is significantly influenced by a cluster of genes on chromosome 5 encoding a family of proteins with enzymatic properties, the 2'-5' oligoadenylate synthetases (OAS). Positional cloning of the locus in question has revealed that susceptibility of laboratory inbred strains to this class of virus is associated with a nonsense mutation in the gene encoding the OAS1B isoform. Analysis of the molecular structure of the cluster in different mammalian species including human indicates that the cluster is extremely polymorphic with a highly variable number of genes and pseudogenes whose functions are not yet completely established. Although still preliminary, a few recent observations also substantiate a possible role for OAS1 in human susceptibility to viral infections (West Nile virus, SARS, etc.) and its possible involvement in some other diseases such as type 1 diabetes and multiple sclerosis. Finally, convergent observations indicate that the molecules encoded by the 2 '-5' OAS cluster might be involved in other fundamental cellular functions such as cell growth and differentiation, gene regulation, and apoptosis.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Flavivirus Infections/immunology , Immunity, Innate , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Flavivirus Infections/etiology , Genetic Predisposition to Disease , Humans , Mice
15.
Mol Immunol ; 45(4): 945-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17854897

ABSTRACT

Aquatic animals, especially filter feeders such as sponges [phylum Porifera], are exposed to a higher viral load than terrestrial species. Until now, the antiviral defense system in the evolutionary oldest multicellular organisms, sponges, is not understood. One powerful protection of vertebrates against virus infection is mediated by the interferon (IFN)-inducible 2'-5'-oligoadenylate synthetase [(2-5)A synthetase] system. In the present study we cloned from the freshwater sponge Lubomirskia baicalensis a cDNA encoding a 314 aa long ORF with a calculated size of 35748Da, a putative (2-5)A synthetase, and raised antibodies against the recombinant protein. The native enzyme was identified in a crude extract from L. baicalensis by application of a novel separation procedure based on polymer coated ferromagnetic nanoparticles. The particles were derivatized with a synthetic double-stranded RNA [dsRNA], synthetic poly(I:C), a known allosteric activator of the latent (2-5)A synthetase. These particles were used to separate a single 35kDa protein from a crude extract of L. baicalensis, which cross-reacted with antibodies raised against the sponge enzyme. In situ hybridization studies revealed that highest expression of the gene is seen in cells surrounding the aquiferous canals. Finally primmorphs, an in vitro cell culture system, from L. baicalensis were exposed to poly(I:C); they responded to this dsRNA with an increased expression of the (2-5)A synthetase gene already after a 1-day incubation period. We conclude that sponges contain the (2-5)A synthetase antiviral protection system.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Porifera/enzymology , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/isolation & purification , Amino Acid Sequence , Animals , Cloning, Molecular , DNA, Complementary/metabolism , Enzyme Activation , In Situ Hybridization , Molecular Sequence Data , Poly I-C/chemistry , Poly I-C/pharmacology , Porifera/cytology , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/pharmacology
16.
Biochimie ; 89(6-7): 779-88, 2007.
Article in English | MEDLINE | ID: mdl-17408844

ABSTRACT

The demonstration by Kerr and colleagues that double-stranded (ds) RNA inhibits drastically protein synthesis in cell-free systems prepared from interferon-treated cells, suggested the existence of an interferon-induced enzyme, which is dependent on dsRNA. Consequently, two distinct dsRNA-dependent enzymes were discovered: a serine/threonine protein kinase that nowadays is referred to as PKR and a 2'-5'oligoadenylate synthetase (2'-5'OAS) that polymerizes ATP to 2'-5'-linked oligomers of adenosine with the general formula pppA(2'p5'A)(n), n>or=1. The product is pppG2'p5'G when GTP is used as a substrate. Three distinct forms of 2'-5'OAS exist in human cells, small, medium, and large, which contain one, two, and three OAS units, respectively, and are encoded by distinct genes clustered on the 2'-5'OAS locus on human chromosome 12. OASL is an OAS like IFN-induced protein encoded by a gene located about 8 Mb telomeric from the 2'-5'OAS locus. OASL is composed of one OAS unit fused at its C-terminus with two ubiquitin-like repeats. The human OASL is devoid of the typical 2'-5'OAS catalytic activity. In addition to these structural differences between the various OAS proteins, the three forms of 2'-5'OAS are characterized by different subcellular locations and enzymatic parameters. These findings illustrate the apparent structural and functional complexity of the human 2'-5'OAS family, and suggest that these proteins may have distinct roles in the cell.


Subject(s)
2',5'-Oligoadenylate Synthetase/chemistry , 2',5'-Oligoadenylate Synthetase/physiology , Interferons/metabolism , Animals , Catalysis , Catalytic Domain , Chromosomes, Human, Pair 12/chemistry , Humans , Interferon alpha-2 , Interferon-alpha/chemistry , Interferons/chemistry , Models, Biological , Models, Genetic , Multigene Family , Protein Structure, Tertiary , RNA, Double-Stranded/chemistry , Recombinant Proteins , Ubiquitin/chemistry
17.
J Immunol ; 178(8): 5166-72, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404299

ABSTRACT

An adenoviral (Ad) vector containing the murine IFN-gamma transgene (Ad:IFN-gamma) was evaluated for its capacity to inhibit HSV-1. To measure effectiveness, viral titers were analyzed in cornea and trigeminal ganglia (TG) during acute ocular HSV-1 infection. Ad:IFN-gamma potently suppressed HSV-1 replication in a dose-dependent fashion, requiring IFN-gamma receptor. Moreover, Ad:IFN-gamma was effective when delivered -72 and -24 h before infection as well as 24 h postinfection. Associated with antiviral opposition, TG from Ad:IFN-gamma-transduced mice harbored fewer T cells. Also related to T cell involvement, Ad:IFN-gamma was effective but attenuated in TG from alphabeta TCR-deficient mice. In corneas, alphabeta TCR(+) T cells were obligatory for protection against viral multiplication. Type I IFN involvement amid antiviral efficacy of Ad:IFN-gamma was further investigated because types I and II IFN pathways have synergistic anti-HSV-1 activity. Ad:IFN-gamma inhibited viral reproduction in corneas and TG from alphabeta IFNR-deficient (CD118(-/-)) mice, although viral titers were 2- to 3-fold higher in cornea and TG compared with wild-type mice. The absence of IFN-stimulated antiviral proteins, 2'-5' oligoadenylate synthetase/RNase L, and dsRNA-dependent protein kinase R completely eliminated the antiviral effectiveness of Ad:IFN-gamma. Collectively, the results demonstrate the following: 1) nonexistence of type I IFN receptor does not abolish defense of Ad:IFN-gamma against HSV-1; 2) antiviral pathways oligoadenylate synthetase-RNase L and protein kinase R are mandatory; and 3) alphabeta TCR(+) T cells are compulsory for Ad:IFN-gamma effectiveness against HSV-1 in cornea but not in TG.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Adenoviridae/genetics , Cornea/virology , Herpesvirus 1, Human/drug effects , Interferon-gamma/pharmacology , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocytes/physiology , eIF-2 Kinase/physiology , Animals , Dose-Response Relationship, Drug , Genetic Vectors , Killer Cells, Natural/physiology , Leukemia Inhibitory Factor Receptor alpha Subunit/physiology , Mice , Mice, Inbred C57BL , Receptors, Interferon/physiology , Trigeminal Ganglion/virology , Interferon gamma Receptor
18.
J Interferon Cytokine Res ; 26(4): 220-5, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16704298

ABSTRACT

Three interferon-gamma (IFN-gamma)-induced antiviral pathways have been reported. Involved antiviral proteins include: Mx, RNase L/2',5'-OAS, and protein kinase R (PKR). Involvement of OAS and PKR in IFN-gamma-induced anti-herpes simplex virus-1 (HSV-1) pathways has not been reported previously, but IFN-gamma induces OAS and PKR when other viruses invade the nervous system. The aim of the current study was to determine if the absence of intact OAS and PKR antiviral pathways affects the antiviral activity of IFN-gamma during acute HSV-1 infection within the trigeminal ganglia (TG). To investigate this, primary TG cultures were established using TGs removed from C57BL/6 (wild-type), RNase L knockout, and RNase L/PKR double knockout mice. Each dissociated TG was transduced with an adenoviral vector containing an IFN-gamma transgene or vector alone. Viral titers after HSV-1 infection of primary TG cell cultures were determined. Significant differences in viral titer for Ad:Null-transduced vs. Ad:IFN-gamma-tranduced TG were found in each genotype. However, the effectiveness of Ad:IFN-gamma was not reduced in the absence of both OAS and PKR pathways or OAS alone. Recombinant IFN-gamma also exhibited anti-HSV-1 activity. The effectiveness of the IFN-gamma transgene was lost in primary TG cells from IFN-gamma receptor knockout mice. The data suggest that novel anti-HSV-1 mechanisms are induced by IFN-gamma.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Herpesvirus 1, Human , Interferon-gamma/genetics , Trigeminal Ganglion/virology , eIF-2 Kinase/physiology , 2',5'-Oligoadenylate Synthetase/genetics , Adenoviridae/genetics , Animals , Cells, Cultured , Genetic Vectors/genetics , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Interferon/genetics , Trigeminal Ganglion/cytology , Trigeminal Ganglion/immunology , eIF-2 Kinase/genetics , Interferon gamma Receptor
19.
J Biol Chem ; 281(8): 4624-37, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16371364

ABSTRACT

The 2',5'-oligoadenylate synthetase (OAS) proteins associated with endoribonuclease RNase L are components of the interferon-regulated OAS/RNase L system, which is an RNA decay pathway known to play an important role in the innate antiviral immunity. A large body of evidence suggests a critical role for the 1b isoform of the mouse Oas gene (Oas1b) in resistance to West Nile virus (WNV) infection in vivo. WNV is a positive, single-stranded RNA virus responsible for severe encephalitis in a large range of animal species and humans. To investigate the molecular basis for the sensitivity of WNV to the Oas1b antiviral pathway, we established a stable mouse fibroblastic cell clone that up-regulates Oas1b protein expression under the control of the Tet-Off expression system. We showed that murine cells respond to Oas1b expression by efficiently inhibiting WNV replication. The antiviral action of Oas1b was essentially restricted to the early stages in virus life cycle. We found that the inability of WNV to productively infect the Oas1b-expressing cells was attributable to a dramatic reduction in positive-stranded viral RNA level. Thus, Oas1b represents an antiviral pathway that exerts its inhibitory effect on WNV replication by preventing viral RNA accumulation inside infected cells.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Antiviral Agents/pharmacology , Virus Replication , West Nile Fever/pathology , West Nile Fever/prevention & control , West Nile virus/drug effects , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Blotting, Western , Cell Line , Cell Survival , Culicidae/virology , DNA Primers/chemistry , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Immunity , Immunoprecipitation , Interferons/metabolism , Mice , Molecular Sequence Data , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Transfection , West Nile virus/genetics
20.
J Immunol ; 175(2): 1100-6, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002711

ABSTRACT

We previously demonstrated that IFN-beta transgene treatment protects mouse trigeminal ganglia (TG) cells from acute HSV-1 infection in vitro. However, IFN-alpha6 transgene treatment does not provide protection against acute HSV-1 infection in vitro, even though equivalent levels of IFN are expressed with both transgene treatments. In the present study we show that IFN-beta transgene treatment before acute ocular HSV-1 infection protects mice from HSV-1-mediated mortality, whereas IFN-alpha6 transgene treatment does not reduce mortality. Treatment with the IFN-beta and IFN-alpha6 transgenes was associated with increased expression of oligoadenylate synthetase (OAS)1a mRNA in the eye. However, protein kinase R mRNA was not up-regulated in the eye. In TG, only IFN-beta transgene treatment reduced infectious virus levels. Furthermore, in the absence of a functional OAS pathway, corneal HSV-1 Ag expression was more widespread, and the ability of IFN-beta transgene treatment to reduce infectious HSV-1 in eyes and TG was lost. Along with selective up-regulation of OAS1a mRNA expression in TG from IFN-beta transgene-treated mice, we found increased levels of phospho-STAT1. Likewise, p38 MAPK phosphorylation was increased in TG from IFN-beta transgene-treated mice, compared with both IFN-alpha6 and vector-treated mice. We also observed a time-dependent increase in JNK phosphorylation in TG from IFN-beta transgene-treated vs IFN-alpha6 and vector-treated mice. Our results demonstrate that IFN-beta is a potent antiviral cytokine that exerts protection against ocular HSV-1 infection via selective up-regulation of OAS1a mRNA in TG and by altering the phosphorylation of proteins in antiviral signaling cascades.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Endoribonucleases/physiology , Herpesvirus 1, Human/immunology , Interferon-beta/physiology , Keratitis, Herpetic/enzymology , Keratitis, Herpetic/immunology , Signal Transduction/immunology , Transgenes , 2',5'-Oligoadenylate Synthetase/biosynthesis , 2',5'-Oligoadenylate Synthetase/deficiency , 2',5'-Oligoadenylate Synthetase/genetics , Acute Disease , Animals , Chlorocebus aethiops , DNA-Binding Proteins/biosynthesis , Endoribonucleases/deficiency , Endoribonucleases/genetics , Interferon-alpha/genetics , Interferon-beta/administration & dosage , Interferon-beta/genetics , Keratitis, Herpetic/mortality , Keratitis, Herpetic/prevention & control , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Phosphorylation , STAT1 Transcription Factor , Signal Transduction/genetics , Trans-Activators/biosynthesis , Transfection , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/virology , Up-Regulation/immunology , Vero Cells , Viral Load , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL