Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Nat Commun ; 12(1): 7166, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887415

ABSTRACT

Programmed -1 ribosomal frameshifting (PRF) in cardioviruses is activated by the 2A protein, a multi-functional virulence factor that also inhibits cap-dependent translational initiation. Here we present the X-ray crystal structure of 2A and show that it selectively binds to a pseudoknot-like conformation of the PRF stimulatory RNA element in the viral genome. Using optical tweezers, we demonstrate that 2A stabilises this RNA element, likely explaining the increase in PRF efficiency in the presence of 2A. Next, we demonstrate a strong interaction between 2A and the small ribosomal subunit and present a cryo-EM structure of 2A bound to initiated 70S ribosomes. Multiple copies of 2A bind to the 16S rRNA where they may compete for binding with initiation and elongation factors. Together, these results define the structural basis for RNA recognition by 2A, show how 2A-mediated stabilisation of an RNA pseudoknot promotes PRF, and reveal how 2A accumulation may shut down translation during virus infection.


Subject(s)
Cardiovirus Infections/virology , Encephalomyocarditis virus/metabolism , Gene Expression Regulation, Viral , Viral Proteins/chemistry , Viral Proteins/metabolism , Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Crystallography, X-Ray , Encephalomyocarditis virus/chemistry , Encephalomyocarditis virus/genetics , Frameshifting, Ribosomal , Humans , Ribosomes/genetics , Ribosomes/metabolism , Viral Proteins/genetics
2.
Int J Mol Sci ; 22(21)2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34768809

ABSTRACT

Virus-induced neurological sequelae resulting from infection by Theiler's murine encephalomyelitis virus (TMEV) are used for studying human conditions ranging from epileptic seizures to demyelinating disease. Mouse strains are typically considered susceptible or resistant to TMEV infection based on viral persistence and extreme phenotypes, such as demyelination. We have identified a broader spectrum of phenotypic outcomes by infecting strains of the genetically diverse Collaborative Cross (CC) mouse resource. We evaluated the chronic-infection gene expression profiles of hippocampi and thoracic spinal cords for 19 CC strains in relation to phenotypic severity and TMEV persistence. Strains were clustered based on similar phenotypic profiles and TMEV levels at 90 days post-infection, and we categorized distinct TMEV response profiles. The three most common profiles included "resistant" and "susceptible," as before, as well as a "resilient" TMEV response group which experienced both TMEV persistence and mild neurological phenotypes even at 90 days post-infection. Each profile had a distinct gene expression signature, allowing the identification of pathways and networks specific to each TMEV response group. CC founder haplotypes for genes involved in these pathways/networks revealed candidate response-specific alleles. These alleles demonstrated pleiotropy and epigenetic (miRNA) regulation in long-term TMEV infection, with particular relevance for resilient mouse strains.


Subject(s)
Cardiovirus Infections/genetics , Gene Expression Regulation , Hippocampus/metabolism , Spinal Cord/metabolism , Theilovirus , Animals , Demyelinating Diseases , Disease Models, Animal , Disease Susceptibility , Female , Male , Mice , Sequence Analysis, RNA
3.
Brain Pathol ; 31(6): e13000, 2021 11.
Article in English | MEDLINE | ID: mdl-34231271

ABSTRACT

Viral infections of the central nervous system cause acute or delayed neuropathology and clinical consequences ranging from asymptomatic courses to chronic, debilitating diseases. The outcome of viral encephalitis is partially determined by genetically programed immune response patterns of the host. Experimental infection of mice with Theiler's murine encephalomyelitis virus (TMEV) causes diverse neurologic diseases, including TMEV-induced demyelinating disease (TMEV-IDD), depending on the used mouse strain. The aim of the present study was to compare initial transcriptomic changes occurring in the brain of TMEV-infected SJL (TMEV-IDD susceptible) and C57BL/6 (TMEV-IDD resistant) mice. Animals were infected with TMEV and sacrificed 4, 7, or 14 days post infection. RNA was isolated from brain tissue and analyzed by whole-transcriptome sequencing. Selected differences were confirmed on a protein level by immunohistochemistry. In mock-infected SJL and C57BL/6 mice, >200 differentially expressed genes (DEGs) were detected. Following TMEV-infection, the number of DEGs increased to >700. Infected C57BL/6 mice showed a higher expression of transcripts related to antigen presentation via major histocompatibility complex (MHC) I, innate antiviral immune responses and cytotoxicity, compared with infected SJL animals. Expression of many of those genes was weaker or delayed in SJL mice, associated with a failure of viral clearance in this mouse strain. SJL mice showed prolonged elevation of MHC II and chemotactic genes compared with C57BL/6 mice, which presumably facilitates the induction of chronic demyelinating disease. In addition, elevated expression of several genes associated with immunomodulatory or -suppressive functions was observed in SJL mice. The exploratory study confirms previous observations in the model and provides an extensive list of new immunologic parameters potentially contributing to different outcomes of viral encephalitis in two mouse strains.


Subject(s)
Brain/metabolism , Cardiovirus Infections/metabolism , Demyelinating Diseases/metabolism , Gene Expression Profiling , Immunity, Innate/physiology , Animals , Brain/pathology , Brain/virology , Cardiovirus Infections/genetics , Cardiovirus Infections/pathology , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Disease Models, Animal , Mice , Theilovirus
4.
PLoS Pathog ; 16(4): e1008457, 2020 04.
Article in English | MEDLINE | ID: mdl-32251420

ABSTRACT

The retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), including RIG-I and melanoma differentiation-associated gene 5 (MDA5), sense cytoplasmic viral RNA and initiate innate antiviral responses. How RIG-I and MDA5 are differentially regulated remains enigmatic. In this study, we identified the guanylate-binding protein (GBP) and zinc-finger FYVE domain-containing protein ZFYVE1 as a negative regulator of MDA5- but not RIG-I-mediated innate antiviral responses. ZFYVE1-deficiency promoted MDA5- but not RIG-I-mediated transcription of downstream antiviral genes. Comparing to wild-type mice, Zfyve1-/- mice were significantly protected from lethality induced by encephalomyocarditis virus (EMCV) that is sensed by MDA5, whereas Zfyve1-/- and Zfyve1+/+ mice were comparable to death induced by vesicular stomatitis virus (VSV) that is sensed by RIG-I. Mechanistically, ZFYVE1 interacted with MDA5 but not RIG-I. ZFYVE1 bound to viral RNA and decreased the ligand binding and oligomerization of MDA5. These findings suggest that ZFYVE1 acts as a specific negative regulator of MDA5-mediated innate immune responses by inhibiting its ligand binding and oligomerization.


Subject(s)
Cardiovirus Infections/immunology , DEAD Box Protein 58/immunology , Encephalomyocarditis virus/physiology , Interferon-Induced Helicase, IFIH1/immunology , Membrane Proteins/immunology , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , DEAD Box Protein 58/genetics , Encephalomyocarditis virus/genetics , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
mBio ; 10(1)2019 02 05.
Article in English | MEDLINE | ID: mdl-30723129

ABSTRACT

Encephalomyocarditis virus (EMCV) is a picornavirus that produces lytic infections in murine and human cells. Employing a genome-wide CRISPR-Cas9 knockout screen to find host factors required for EMCV infection, we identified a role for ADAM9 in EMCV infection. CRISPR-mediated deletion of ADAM9 in multiple human cell lines rendered the cells highly resistant to EMCV infection and cell death. Primary fibroblasts from ADAM9 KO mice were also strongly resistant to EMCV infection and cell death. In contrast, ADAM9 KO and WT cells were equally susceptible to infection with other viruses, including the picornavirus Coxsackie virus B. ADAM9 KO cells failed to produce viral progeny when incubated with EMCV. However, bypassing EMCV entry into cells through delivery of viral RNA directly to the cytosol yielded infectious EMCV virions from ADAM9 KO cells, suggesting that ADAM9 is not required for EMCV replication post-entry. These findings establish that ADAM9 is required for the early stage of EMCV infection, likely for virus entry or viral genome delivery to the cytosol.IMPORTANCE Viral myocarditis is a leading cause of death in the United States, contributing to numerous unexplained deaths in people ≤35 years old. Enteroviruses contribute to many cases of human myocarditis. Encephalomyocarditis virus (EMCV) infection causes viral myocarditis in rodent models, but its receptor requirements have not been fully identified. CRISPR-Cas9 screens can identify host dependency factors essential for EMCV infection and enhance our understanding of key events that follow viral infection, potentially leading to new strategies for preventing viral myocarditis. Using a CRISPR-Cas9 screen, we identified adisintegrin and metalloproteinase 9 domain (ADAM9) as a major factor required for the early stages of EMCV infection in both human and murine infection.


Subject(s)
ADAM Proteins/metabolism , Cardiovirus Infections/genetics , Disease Resistance , Encephalomyocarditis virus/growth & development , Membrane Proteins/metabolism , Animals , Cell Line , Gene Knockout Techniques , Genetic Testing , Humans , Mice , Mice, Knockout , Models, Biological
6.
PLoS Pathog ; 14(10): e1007307, 2018 10.
Article in English | MEDLINE | ID: mdl-30308076

ABSTRACT

As antimicrobial signalling molecules, type III or lambda interferons (IFNλs) are critical for defence against infection by diverse pathogens, including bacteria, fungi and viruses. Counter-intuitively, expression of one member of the family, IFNλ4, is associated with decreased clearance of hepatitis C virus (HCV) in the human population; by contrast, a natural frameshift mutation that abrogates IFNλ4 production improves HCV clearance. To further understand how genetic variation between and within species affects IFNλ4 function, we screened a panel of all known extant coding variants of human IFNλ4 for their antiviral potential and identify three that substantially affect activity: P70S, L79F and K154E. The most notable variant was K154E, which was found in African Congo rainforest 'Pygmy' hunter-gatherers. K154E greatly enhanced in vitro activity in a range of antiviral (HCV, Zika virus, influenza virus and encephalomyocarditis virus) and gene expression assays. Remarkably, E154 is the ancestral residue in mammalian IFNλ4s and is extremely well conserved, yet K154 has been fixed throughout evolution of the hominid genus Homo, including Neanderthals. Compared to chimpanzee IFNλ4, the human orthologue had reduced activity due to amino acid K154. Comparison of published gene expression data from humans and chimpanzees showed that this difference in activity between K154 and E154 in IFNλ4 correlates with differences in antiviral gene expression in vivo during HCV infection. Mechanistically, our data show that the human-specific K154 negatively affects IFNλ4 activity through a novel means by reducing its secretion and potency. We thus demonstrate that attenuated activity of IFNλ4 is conserved among humans and postulate that differences in IFNλ4 activity between species contribute to distinct host-specific responses to-and outcomes of-infection, such as HCV infection. The driver of reduced IFNλ4 antiviral activity in humans remains unknown but likely arose between 6 million and 360,000 years ago in Africa.


Subject(s)
Antiviral Agents/therapeutic use , Cardiovirus Infections/drug therapy , Hepatitis C/drug therapy , Interleukins/genetics , Polymorphism, Single Nucleotide , Zika Virus Infection/drug therapy , Animals , Biological Evolution , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cells, Cultured , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/isolation & purification , Gene Expression Regulation , Hepacivirus/drug effects , Hepacivirus/isolation & purification , Hepatitis C/genetics , Hepatitis C/virology , Humans , Pan troglodytes , Species Specificity , Zika Virus/drug effects , Zika Virus/isolation & purification , Zika Virus Infection/genetics , Zika Virus Infection/virology
7.
Sci Rep ; 8(1): 6106, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29666403

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) infection represents an experimental mouse model to study hippocampal damage induced by neurotropic viruses. IL-10 is a pleiotropic cytokine with profound anti-inflammatory properties, which critically controls immune homeostasis. In order to analyze IL-10R signaling following virus-induced polioencephalitis, SJL mice were intracerebrally infected with TMEV. RNA-based next generation sequencing revealed an up-regulation of Il10, Il10rα and further genes involved in IL-10 downstream signaling, including Jak1, Socs3 and Stat3 in the brain upon infection. Subsequent antibody-mediated blockade of IL-10R signaling led to enhanced hippocampal damage with neuronal loss and increased recruitment of CD3+ T cells, CD45R+ B cells and an up-regulation of Il1α mRNA. Increased expression of Tgfß and Foxp3 as well as accumulation of Foxp3+ regulatory T cells and arginase-1+ macrophages/microglia was detected in the hippocampus, representing a potential compensatory mechanism following disturbed IL-10R signaling. Additionally, an increased peripheral Chi3l3 expression was found in spleens of infected mice, which may embody reactive regulatory mechanisms for prevention of excessive immunopathology. The present study highlights the importance of IL-10R signaling for immune regulation and its neuroprotective properties in the context of an acute neurotropic virus infection.


Subject(s)
Cardiovirus Infections/immunology , Hippocampus/immunology , Receptors, Interleukin-10/immunology , Theilovirus/immunology , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , Female , Hippocampus/pathology , Hippocampus/virology , Mice, Inbred C57BL , Receptors, Interleukin-10/genetics , Signal Transduction , Up-Regulation
8.
PLoS Pathog ; 14(2): e1006886, 2018 02.
Article in English | MEDLINE | ID: mdl-29462185

ABSTRACT

Melanoma differentiation-associated gene-5 (MDA5) recognizes distinct subsets of viruses including Encephalomyocarditis virus (EMCV) of picornavirus family, but the molecular mechanisms underlying the specificity of the viral recognition of MDA5 in immune cells remain obscure. DHX29 is an RNA helicase required for the translation of 5' structured mRNA of host and many picornaviruses (such as EMCV). We identify that DXH29 as a key RNA co-sensor, plays a significant role for specific recognition and triggering anti-EMCV immunity. We have observed that DHX29 regulates MDA5-, but not RIG-I-, mediated type I interferon signaling by preferentially interacting with structured RNAs and specifically with MDA5 for enhancing MDA5-dsRNA binding affinity. Overall, our results identify a critical role for DHX29 in innate immune response and provide molecular insights into the mechanisms by which DHX29 recognizes 5' structured EMCV RNA and interacts with MDA5 for potent type I interferon signaling and antiviral immunity.


Subject(s)
Cardiovirus Infections/immunology , Encephalomyocarditis virus/immunology , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1/physiology , RNA Helicases/physiology , RNA, Viral/immunology , Animals , Cardiovirus Infections/genetics , Cells, Cultured , Chlorocebus aethiops , Encephalomyocarditis virus/genetics , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , RNA Helicases/genetics , RNA, Viral/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Vero Cells
9.
Virology ; 516: 139-146, 2018 03.
Article in English | MEDLINE | ID: mdl-29353210

ABSTRACT

Cardioviruses cause diseases in many animals including, in rare cases, humans. Although they share common features with all picornaviruses, cardioviruses have unique properties that distinguish them from other family members, including enteroviruses. One feature shared by all picornaviruses is the covalent attachment of VPg to the 5' end of genomic RNA via a phosphotyrosyl linkage. For enteroviruses, this linkage is cleaved by a host cell protein, TDP2. Since TDP2 is divergently required during enterovirus infections, we determined if TDP2 is necessary during infection by the prototype cardiovirus, EMCV. We found that EMCV yields are reduced in the absence of TDP2. We observed a decrease in viral protein accumulation and viral RNA replication in the absence of TDP2. In contrast to enterovirus infections, we found that TDP2 is modified at peak times of EMCV infection. This finding suggests a unique mechanism for cardioviruses to regulate TDP2 activity during infection.


Subject(s)
Cardiovirus Infections/metabolism , Cardiovirus/metabolism , Nuclear Proteins/metabolism , Phosphoric Diester Hydrolases/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Viral Proteins/metabolism , Animals , Cardiovirus/genetics , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , DNA-Binding Proteins , Fibroblasts/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Mice , Mice, Knockout , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Protein Transport , Proteolysis , RNA, Viral/genetics , RNA, Viral/metabolism , Transcription Factors/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Viral Proteins/genetics , Virus Replication
10.
Biochem Biophys Res Commun ; 494(1-2): 227-233, 2017 12 09.
Article in English | MEDLINE | ID: mdl-29032202

ABSTRACT

Laboratory of genetics and physiology 2 (LGP2) and melanoma differentiation-associated gene 5 (MDA5) cooperatively detect viral RNA in the cytoplasm of Cardiovirus-infected cells and activate innate immune responses. Here, we evaluated whether the double-stranded RNA-binding protein PACT plays a role in this anti-viral response to further elucidate the mechanism. Immunoprecipitation experiments demonstrated that PACT interacts with LGP2 and that this interaction is enhanced by encephalomyocarditis virus (EMCV) infection. In vitro interaction analyses using purified recombinant proteins confirmed that the single-stranded Theiler's murine encephalitis virus genome enhanced the interaction between LGP2 and PACT. Small interfering RNA knockdown experiments further indicated that PACT is required for Cardiovirus-triggered interferon responses. To support this functional interaction with LGP2, overexpressed PACT was shown to enhance EMCV-triggered interferon promoter activity only when LGP2 and MDA5 were co-expressed but not when MDA5 is expressed alone. Together, our findings indicate a possible role of PACT in regulating the Cardiovirus-triggered immune responses mediated by MDA5 and LGP2, which opens the door to novel therapeutic strategies in interferon-related autoimmune diseases and cancer.


Subject(s)
Cardiovirus Infections/immunology , Encephalomyocarditis virus , Interferon-Induced Helicase, IFIH1/immunology , RNA Helicases/immunology , RNA-Binding Proteins/immunology , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cell Line , Chlorocebus aethiops , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Gene Knockdown Techniques , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/genetics , Mice , Promoter Regions, Genetic , RNA Helicases/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , RNA, Viral/immunology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Ribonuclease III/antagonists & inhibitors , Ribonuclease III/genetics , Ribonuclease III/immunology , Vero Cells
11.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28916264

ABSTRACT

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , RNA Transport , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cell Line , Cytoplasm , DEAD Box Protein 58/metabolism , Disease Models, Animal , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Endosomes/metabolism , Female , Gene Expression , Gene Knockout Techniques , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Nucleotide Transport Proteins , Protein Binding , Protein Transport , RNA, Viral/genetics , RNA, Viral/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism
12.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553952

ABSTRACT

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Subject(s)
Autoimmunity/genetics , Cardiovirus Infections/genetics , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/genetics , Adolescent , Adult , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/immunology , Blotting, Southern , Cardiovirus Infections/immunology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Encephalomyocarditis virus/immunology , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Immunoblotting , Interferon-Induced Helicase, IFIH1/immunology , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Diseases/genetics , Virus Diseases/immunology , Young Adult
13.
Proc Natl Acad Sci U S A ; 113(37): 10388-93, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27582466

ABSTRACT

Despite the breadth of knowledge that exists regarding the function of long noncoding RNAs (lncRNAs) in biological phenomena, the role of lncRNAs in host antiviral responses is poorly understood. Here, we report that lncRNA#32 is associated with type I IFN signaling. The silencing of lncRNA#32 dramatically reduced the level of IFN-stimulated gene (ISG) expression, resulting in sensitivity to encephalomyocarditis virus (EMCV) infection. In contrast, the ectopic expression of lncRNA#32 significantly suppressed EMCV replication, suggesting that lncRNA#32 positively regulates the host antiviral response. We further demonstrated the suppressive function of lncRNA#32 in hepatitis B virus and hepatitis C virus infection. lncRNA#32 bound to activating transcription factor 2 (ATF2) and regulated ISG expression. Our results reveal a role for lncRNA#32 in host antiviral responses.


Subject(s)
Activating Transcription Factor 2/genetics , Host-Pathogen Interactions/genetics , Interferon Type I/genetics , RNA, Long Noncoding/genetics , Activating Transcription Factor 2/metabolism , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cell Line, Tumor , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/pathogenicity , Gene Expression Regulation , Gene Silencing , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepatitis C/genetics , Hepatitis C/virology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon Type I/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Virus Replication/genetics
14.
Exp Mol Pathol ; 100(3): 434-40, 2016 06.
Article in English | MEDLINE | ID: mdl-27079771

ABSTRACT

The present study was carried out to clarify the mechanisms of EMC virus-induced sialodacryoadenitis in mice during the acute phase infection focusing on the activation of type I interferon (IFN) signaling in the parotid and exorbital lachrymal glands. In the parotid gland, a few apoptotic acinar cells were detected at 2days post inoculation (DPI). The ratio of apoptotic acinar cells increased at 3 and 4DPI. On the other hand, in the exorbital lachrymal gland, apoptosis of acinar cells and infiltration of inflammatory cells mainly composed of mononuclear cells started at 3DPI, and prominent acinar cell damage developed at 4DPI. Viral RNA was detected at 3 and 4DPI in both glands and the expression level was higher in the exorbital lachrymal gland than in the parotid gland. The up-regulation of IFN-stimulated genes (ISGs), such as Irf7, Pkr and Oas, was quickly induced at 2DPI in the parotid gland, and this probably contributed to suppress viral replication and to eliminate affected cells by apoptosis. In the exorbital lachrymal gland, the expression levels of ISGs mRNAs were not elevated at 2DPI, suggesting no induction of an effective anti-viral response such as apoptosis at this time point. In the exorbital lachrymal gland, the mRNA expression of IFN beta and IFN alpha (type I IFNs) was weak- to strong-positive at 1DPI, and became negative at 2DPI. The weak- to strong-positive expression of IFNs at 1DPI is likely related to the abrupt viral replication and pathological changes in the exorbital lachrymal gland through activating the negative feedback regulation that depressed the IFN signaling cascade at 2DPI. In conclusion, the present study showed the changes in factors involved in the activation of type I IFN signaling cascade in the parotid and exorbital lachrymal glands and their differences between the two glands during the acute phase of EMC virus infection in mice.


Subject(s)
Cardiovirus Infections/metabolism , Interferon Type I/genetics , Lacrimal Apparatus/metabolism , Parotid Gland/metabolism , Signal Transduction/genetics , Acute Disease , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/physiology , Gene Expression , Host-Pathogen Interactions , Interferon Regulatory Factor-7/genetics , Interferon-alpha/genetics , Lacrimal Apparatus/virology , Male , Mice, Inbred DBA , Parotid Gland/virology , RNA, Viral/genetics , RNA, Viral/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
15.
PLoS One ; 11(2): e0148184, 2016.
Article in English | MEDLINE | ID: mdl-26828718

ABSTRACT

OBJECTIVE: Saffold virus (SAFV), a picornavirus, is occasionally detected in children with acute flaccid paralysis, meningitis, and cerebellitis; however, the neuropathogenicity of SAFV remains undetermined. METHODS: The virulence of two clinical isolates of SAFV type 3 (SAFV-3) obtained from a patient with aseptic meningitis (AM strain) and acute upper respiratory inflammation (UR strain) was analyzed in neonatal and young mice utilizing virological, pathological, and immunological methods. RESULTS: The polyproteins of the strains differed in eight amino acids. Both clinical isolates were infective, exhibited neurotropism, and were mildly neurovirulent in neonatal ddY mice. Both strains pathologically infected neural progenitor cells and glial cells, but not large neurons, with the UR strain also infecting epithelial cells. UR infection resulted in longer inflammation in the brain and spinal cord because of demyelination, while the AM strain showed more infectivity in the cerebellum in neonatal ddY mice. Additionally, young BALB/c mice seroconverted following mucosal inoculation with the UR, but not the AM, strain. CONCLUSIONS: Both SAFV-3 isolates had neurotropism and mild neurovirulence but showed different cell tropisms in both neonatal and young mouse models. This animal model has the potential to recapitulate the potential neuropathogenicity of SAFV-3.


Subject(s)
Brain/pathology , Brain/virology , Cardiovirus Infections/virology , Cardiovirus/isolation & purification , Cardiovirus/pathogenicity , Amino Acid Sequence , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Body Weight , Cardiovirus/immunology , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cardiovirus Infections/pathology , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Female , Immunity , Inflammation/pathology , Injections, Intraventricular , Interferon Type I/metabolism , Mice, Inbred BALB C , Mucous Membrane/pathology , Mucous Membrane/virology , Real-Time Polymerase Chain Reaction , Tropism , Virulence , Virus Replication
16.
J Neurovirol ; 22(1): 66-79, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26260496

ABSTRACT

Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) is an important model of the progressive disability caused by irreversible CNS tissue injury, and provides an example of how a CNS pathogen can cause inflammation, demyelination, and neuronal damage. We were interested in which molecules, especially inflammatory mediators, might be upregulated in the CNS throughout TMEV-IDD. We quantitated by a real-time RT-PCR multi-gene system the expression of a pathway-focused panel of genes at 30 and 165 days post infection, characterizing both the early inflammatory and the late neurodegenerative stages of TMEV-IDD. Also, we measured 32 cytokines/chemokines by multiplex Luminex analysis in CSF specimens from early and late TMEV-IDD as well as sham-treated mice. Results indicate that, in the later stage of TMEV-IDD, activation of the innate immune response is most prominent: TLRs, type I IFN response genes, and innate immunity-associated cytokines were highly expressed in late TMEV-IDD compared to sham (p ≤ 0.0001) and early TMEV-IDD (p < 0.05). Conversely, several molecular mediators of adaptive immune response were highly expressed in early TMEV-IDD (all p ≤ 0.001). Protein detection in the CSF was broadly concordant with mRNA abundance of the corresponding gene measured by real-time RT-PCR in the spinal cord, since several cytokines/chemokines were increased in the CSF of TMEV-IDD mice. Results show a clear shift from adaptive to innate immunity from early to late TMEV-IDD, indicating that adaptive and innate immune pathways are likely involved in the development and progression of the disease to different extents. CSF provides an optimal source of biomarkers of CNS neuroinflammation.


Subject(s)
Adaptive Immunity , Cardiovirus Infections/immunology , Demyelinating Diseases/immunology , Host-Pathogen Interactions , Immunity, Innate , Animals , Cardiovirus Infections/cerebrospinal fluid , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Central Nervous System/immunology , Central Nervous System/virology , Cytokines/cerebrospinal fluid , Cytokines/genetics , Cytokines/immunology , Demyelinating Diseases/cerebrospinal fluid , Demyelinating Diseases/genetics , Demyelinating Diseases/virology , Disease Progression , Gene Expression Regulation , Inflammation , Mice , Molecular Sequence Annotation , RNA, Messenger/cerebrospinal fluid , RNA, Messenger/genetics , RNA, Messenger/immunology , Real-Time Polymerase Chain Reaction , Theilovirus/growth & development , Theilovirus/immunology , Theilovirus/pathogenicity , Time Factors , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
17.
Nat Commun ; 6: 6748, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25849081

ABSTRACT

Accumulating evidence suggests that viruses play an important role in the development of diabetes. Although the diabetogenic encephalomyocarditis strain D virus induces diabetes in restricted lines of inbred mice, the susceptibility genes to virus-induced diabetes have not been identified. We report here that novel Tyrosine kinase 2 (Tyk2) gene mutations are present in virus-induced diabetes-sensitive SJL and SWR mice. Mice carrying the mutant Tyk2 gene on the virus-resistant C57BL/6 background are highly sensitive to virus-induced diabetes. Tyk2 gene expression is strongly reduced in Tyk2-mutant mice, associated with low Tyk2 promoter activity, and leads to decreased expression of interferon-inducible genes, resulting in significantly compromised antiviral response. Tyk2-mutant pancreatic ß-cells are unresponsive even to high dose of Type I interferon. Reversal of virus-induced diabetes could be achieved by ß-cell-specific Tyk2 gene expression. Thus, reduced Tyk2 gene expression in pancreatic ß-cells due to natural mutation is responsible for susceptibility to virus-induced diabetes.


Subject(s)
Cardiovirus Infections/genetics , Diabetes Mellitus, Experimental/genetics , Encephalomyocarditis virus , Insulin-Secreting Cells/metabolism , RNA, Messenger/metabolism , TYK2 Kinase/genetics , Animals , Diabetes Mellitus, Experimental/virology , Gene Expression , Genetic Predisposition to Disease , Interferon Type I , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mutation , TYK2 Kinase/metabolism
18.
J Immunol ; 194(6): 2796-809, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681345

ABSTRACT

Virus-induced myositis is an emerging global affliction that remains poorly characterized with few treatment options. Moreover, muscle-tropic viruses often spread to the CNS, causing dramatically increased morbidity. Therefore, there is an urgent need to explore genetic factors involved in this class of human disease. This report investigates critical innate immune pathways affecting murine virus-induced myositis. Of particular importance, the key immune regulator src homology region 2 domain-containing phosphatase 1 (SHP-1), which normally suppresses macrophage-mediated inflammation, is a major factor in promoting clinical disease in muscle. We show that Theiler's murine encephalomyelitis virus (TMEV) infection of skeletal myofibers induces inflammation and subsequent dystrophic calcification, with loss of ambulation in wild-type (WT) mice. Surprisingly, although similar extensive myofiber infection and inflammation are observed in SHP-1(-/-) mice, these mice neither accumulate dead calcified myofibers nor lose ambulation. Macrophages were the predominant effector cells infiltrating WT and SHP-1(-/-) muscle, and an increased infiltration of immature monocytes/macrophages correlated with an absence of clinical disease in SHP-1(-/-) mice, whereas mature M1-like macrophages corresponded with increased myofiber degeneration in WT mice. Furthermore, blocking SHP-1 activation in WT macrophages blocked virus-induced myofiber degeneration, and pharmacologic ablation of macrophages inhibited muscle calcification in TMEV-infected WT animals. These data suggest that, following TMEV infection of muscle, SHP-1 promotes M1 differentiation of infiltrating macrophages, and these inflammatory macrophages are likely involved in damaging muscle fibers. These findings reveal a pathological role for SHP-1 in promoting inflammatory macrophage differentiation and myofiber damage in virus-infected skeletal muscle, thus identifying SHP-1 and M1 macrophages as essential mediators of virus-induced myopathy.


Subject(s)
Cardiovirus Infections/immunology , Cell Differentiation/immunology , Macrophages/immunology , Myositis/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Theilovirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Cell Differentiation/genetics , Flow Cytometry , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/virology , Myositis/genetics , Myositis/virology , Oligonucleotide Array Sequence Analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Theilovirus/physiology , Transcriptome/immunology , Virus Replication/immunology
19.
J Neurovirol ; 20(5): 485-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052192

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) induces demyelination in susceptible strains of mice (SJL/J) through an immunopathological process that is mediated by CD4(+) Th1 T cell. These T cells are chemoattracted to the central nervous system by chemokines. Hence, in this study, we focused on the production of the chemokine "interferon-gamma-inducible protein 10 kDa," or IP-10/CXCL10, by cultured SJL/J mouse astrocytes infected with the BeAn strain of TMEV and its capacity to attract activated T cells. The analysis of the whole murine genome by DNA hybridization with cRNAs from mock- and TMEV-infected cultures revealed the upregulation of six sequences that potentially encode for CXCL10. This increased CXCL10 expression was validated by PCR and qPCR. The presence of this chemokine was further demonstrated by enzyme-linked immunoassay (ELISA). Significantly, astrocytes from BALB/c mice, a strain resistant to demyelination, did not produce CXCL10. The secreted CXCL10 was biologically active, inducing chemoattraction of activated lymphocytes. The inflammatory cytokines, IL-1α, IFN-γ, and TNF-α, were strong inducers of CXCL10 in astrocytes. Serum from TMEV-infected SJL/J but not BALB/c mice contains CXCL10, the levels of which peak at the onset of the clinical disease. Finally, this in vitro inflammation model was fully inhibited by 17ß-estradiol and four selective estrogen receptor modulators, as demonstrated by ELISA and qPCR.


Subject(s)
Astrocytes/virology , Cardiovirus Infections/immunology , Chemokine CXCL10/biosynthesis , T-Lymphocytes/immunology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Cells, Cultured , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , In Situ Hybridization , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Selective Estrogen Receptor Modulators/pharmacology , Theilovirus/immunology , Up-Regulation
20.
J Virol ; 88(18): 10748-57, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25008915

ABSTRACT

UNLABELLED: Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are essential intracellular detectors of viral RNA. They contribute to the type I interferon (IFN) response that is crucial for host defense against viral infections. Given the potent antiviral and proinflammatory activities elicited by the type I IFNs, induction of the type I IFN response is tightly regulated. Members of the tripartite motif (TRIM) family of proteins have recently emerged as key regulators of antiviral immunity. We show that TRIM13, an E3 ubiquitin ligase, is expressed in immune cells and is upregulated in bone marrow-derived macrophages upon stimulation with inducers of type I IFN. TRIM13 interacts with MDA5 and negatively regulates MDA5-mediated type I IFN production in vitro, acting upstream of IFN regulatory factor 3. We generated Trim13(-/-) mice and show that upon lethal challenge with encephalomyocarditis virus (EMCV), which is sensed by MDA5, Trim13(-/-) mice produce increased amounts of type I IFNs and survive longer than wild-type mice. Trim13(-/-) murine embryonic fibroblasts (MEFs) challenged with EMCV or poly(I · C) also show a significant increase in beta IFN (IFN-ß) levels, but, in contrast, IFN-ß responses to the RIG-I-detected Sendai virus were diminished, suggesting that TRIM13 may play a role in positively regulating RIG-I function. Together, these results demonstrate that TRIM13 regulates the type I IFN response through inhibition of MDA5 activity and that it functions nonredundantly to modulate MDA5 during EMCV infection. IMPORTANCE: The type I interferon (IFN) response is crucial for host defense against viral infections, and proper regulation of this pathway contributes to maintaining immune homeostasis. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are intracellular detectors of viral RNA that induce the type I IFN response. In this study, we show that expression of the gene tripartite motif 13 (Trim13) is upregulated in response to inducers of type I IFN and that TRIM13 interacts with both MDA5 and RIG-I in vitro. Through the use of multiple in vitro and in vivo model systems, we show that TRIM13 is a negative regulator of MDA5-mediated type I IFN production and may also impact RIG-I-mediated type I IFN production by enhancing RIG-I activity. This places TRIM13 at a key junction within the viral response pathway and identifies it as one of the few known modulators of MDA5 activity.


Subject(s)
Cardiovirus Infections/enzymology , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Encephalomyocarditis virus/physiology , Interferon-alpha/metabolism , Interferon-beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Cardiovirus Infections/virology , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1 , Interferon-alpha/genetics , Interferon-beta/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...