Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
mBio ; 12(3)2021 05 11.
Article in English | MEDLINE | ID: mdl-33975932

ABSTRACT

Intestinal microbiomes are of vital importance in antagonizing systemic viral infection. However, very little literature has shown whether commensal bacteria play a crucial role in protecting against enteric virus systemic infection from the aspect of modulating host innate immunity. In the present study, we utilized an enteric virus, encephalomyocarditis virus (EMCV), to inoculate mice treated with phosphate-buffered saline (PBS) or given an antibiotic cocktail (Abx) orally or intraperitoneally to examine the impact of microbiota depletion on virulence and viral replication in vivo Microbiota depletion exacerbated the mortality, neuropathogenesis, viremia, and viral burden in brains following EMCV infection. Furthermore, Abx-treated mice exhibited severely diminished mononuclear phagocyte activation and impaired type I interferon (IFN) production and expression of IFN-stimulated genes (ISG) in peripheral blood mononuclear cells (PBMC), spleens, and brains. With the help of fecal bacterial 16S rRNA sequencing of PBS- and Abx-treated mice, we identified a single commensal bacterium, Blautia coccoides, that can restore mononuclear phagocyte- and IFNAR (IFN-α/ß receptor)-dependent type I IFN responses to restrict systemic enteric virus infection. These findings may provide insight into the development of novel therapeutics for preventing enteric virus infection or possibly alleviating clinical diseases by activating host systemic innate immune responses via respective probiotic treatment using B. coccoidesIMPORTANCE While cumulative data indicate that indigenous commensal bacteria can facilitate enteric virus infection, little is known regarding whether intestinal microbes have a protective role in antagonizing enteric systemic infection by modulating host innate immunity. Although accumulating literature has pointed out that the microbiota has a fundamental impact on host systemic antiviral innate immune responses mediated by type I interferon (IFN), only a few specific commensal bacteria species have been revealed to be capable of regulating IFN-I and ISG expression, not to mention the underlying mechanisms. Thus, it is important to understand the cross talk between microbiota and host anti-enteric virus innate immune responses and characterize the specific bacterial species that possess protective functions. Our study demonstrates how fundamental innate immune mediators such as mononuclear phagocytes and type I IFN are regulated by commensal bacteria to antagonize enteric virus systemic infection. In particular, we have identified a novel commensal bacterium, Blautia coccoides, that can restrict enteric virus replication and neuropathogenesis by activating IFN-I and ISG responses in mononuclear phagocytes via an IFNAR- and STAT1-mediated signaling pathway.


Subject(s)
Cardiovirus Infections/prevention & control , Encephalomyocarditis virus/immunology , Gastrointestinal Microbiome/immunology , Immunity, Innate , Interferon Type I/immunology , Viremia/immunology , Viremia/prevention & control , Animals , Anti-Bacterial Agents/administration & dosage , Cardiovirus Infections/immunology , Clostridiales/immunology , Encephalomyocarditis virus/pathogenicity , Female , Gastrointestinal Microbiome/drug effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Symbiosis/immunology , Virus Replication/immunology
2.
Sci Rep ; 11(1): 6331, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737691

ABSTRACT

Few publications, often limited to one specific pathogen, have studied bonobos (Pan paniscus), our closest living relatives, as possible reservoirs of certain human infectious agents. Here, 91 stool samples from semicaptive bonobos and bonobos reintroduced in the wild, in the Democratic Republic of the Congo, were screened for different infectious agents: viruses, bacteria and parasites. We showed the presence of potentially zoonotic viral, bacterial or parasitic agents in stool samples, sometimes coinfecting the same individuals. A high prevalence of Human mastadenoviruses (HAdV-C, HAdV-B, HAdV-E) was observed. Encephalomyocarditis viruses were identified in semicaptive bonobos, although identified genotypes were different from those identified in the previous fatal myocarditis epidemic at the same site in 2009. Non-pallidum Treponema spp. including symbiotic T. succinifaciens, T. berlinense and several potential new species with unknown pathogenicity were identified. We detected DNA of non-tuberculosis Mycobacterium spp., Acinetobacter spp., Salmonella spp. as well as pathogenic Leptospira interrogans. Zoonotic parasites such as Taenia solium and Strongyloides stercoralis were predominantly present in wild bonobos, while Giardia lamblia was found only in bonobos in contact with humans, suggesting a possible exchange. One third of bonobos carried Oesophagostomum spp., particularly zoonotic O. stephanostomum and O. bifurcum-like species, as well as other uncharacterized Nematoda. Trypanosoma theileri has been identified in semicaptive bonobos. Pathogens typically known to be transmitted sexually were not identified. We present here the results of a reasonably-sized screening study detecting DNA/RNA sequence evidence of potentially pathogenic viruses and microorganisms in bonobo based on a noninvasive sampling method (feces) and focused PCR diagnostics.


Subject(s)
Endangered Species , Host-Pathogen Interactions/genetics , Mastadenovirus/isolation & purification , Pan paniscus/virology , Animals , Democratic Republic of the Congo/epidemiology , Encephalomyocarditis virus/isolation & purification , Encephalomyocarditis virus/pathogenicity , Feces/microbiology , Feces/parasitology , Feces/virology , Humans , Mastadenovirus/pathogenicity , Pan paniscus/microbiology , Pan paniscus/parasitology , Pan troglodytes/microbiology , Pan troglodytes/parasitology , Pan troglodytes/virology , Parasites/isolation & purification , Parasites/pathogenicity
3.
J Gen Virol ; 102(3)2021 03.
Article in English | MEDLINE | ID: mdl-33533710

ABSTRACT

Encephalomyocarditis virus (EMCV) infects a wide range of hosts and can cause encephalitis, myocarditis, reproductive disorders and diabetes mellitus in selected mammalian species. As for humans, EMCV infection seems to occur by the contact with animals and can cause febrile illnesses in some infected patients. Here we isolated EMCV strain ZM12/14 from a natal multimammate mouse (Mastomys natalensis: M. natalensis) in Zambia. Pairwise sequence similarity of the ZM12/14 P1 region consisting of antigenic capsid proteins showed the highest similarity of nucleotide (80.7 %) and amino acid (96.2%) sequence with EMCV serotype 1 (EMCV-1). Phylogenetic analysis revealed that ZM12/14 clustered into EMCV-1 at the P1 and P3 regions but segregated from known EMCV strains at the P2 region, suggesting a unique evolutionary history. Reverse transcription PCR (RT-PCR) screening and neutralizing antibody assays for EMCV were performed using collected tissues and serum from various rodents (n=179) captured in different areas in Zambia. We detected the EMCV genome in 19 M. natalensis (19/179=10.6 %) and neutralizing antibody for EMCV in 33 M. natalensis (33/179=18.4 %). However, we did not detect either the genome or neutralizing antibody in other rodent species. High neutralizing antibody litres (≧320) were observed in both RT-PCR-negative and -positive animals. Inoculation of ZM12/14 caused asymptomatic persistent infection in BALB/c mice with high antibody titres and high viral loads in some organs, consistent with the above epidemiological results. This study is the first report of the isolation of EMCV in Zambia, suggesting that M. natalensis may play a role as a natural reservoir of infection.


Subject(s)
Cardiovirus Infections/veterinary , Disease Reservoirs/virology , Encephalomyocarditis virus/isolation & purification , Murinae/virology , Rodent Diseases/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cardiovirus Infections/epidemiology , Cardiovirus Infections/virology , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Encephalomyocarditis virus/pathogenicity , Evolution, Molecular , Genome, Viral , Mice, Inbred BALB C , Phylogeny , Prevalence , Rodent Diseases/epidemiology , Shrews/virology , Zambia/epidemiology
4.
Virus Genes ; 55(6): 786-794, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31367998

ABSTRACT

Foot-and-mouth disease (FMD) is a host-restricted disease of cloven-hoofed animals, such as cattle and pigs. There are seven major serotypes of FMD virus that exhibit high antigenic variation, making vaccine strain selection difficult. However, there is an internal ribosomal entry site (IRES) element within the 5' untranslated region of the FMD virus (FMDV) RNA genome that is relatively conserved among FMDV serotypes and could be used as a pan-serotype target for disease interventions. To determine the potential for targeting the IRES as promising drug target, we designed a short interfering RNA (siRNA) targeting a relatively conserved region in the FMDV-IRES. The siRNA affected FMDV-IRES expression but not the expression of the encephalomyocarditis virus or hepatitis C virus IRES. To evaluate the effects of siRNA-mediated silencing, we established cell lines expressing a bicistronic luciferase reporter plasmid, which contained an FMDV-IRES element between the Renilla and firefly luciferase genes. The designed siRNA inhibited FMDV-IRES-mediated translation in a concentration-dependent manner. In order to sustain this inhibitory effect, we designed a short hairpin RNA (shRNA)-expressing lentiviral vector. The results showed that the lenti-shRNA vector significantly suppressed FMDV-IRES activity for up to 2 weeks in cell culture. Thus, our findings in this study provided a basis for the development of effective pan-serotype FMDV inhibitors.


Subject(s)
Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease/genetics , Internal Ribosome Entry Sites/genetics , Virus Replication/genetics , Animals , Cattle , Cell Line , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/pathogenicity , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/pathogenicity , Gene Expression Regulation, Viral/genetics , Gene Silencing , Hepacivirus/genetics , Hepacivirus/pathogenicity , RNA, Small Interfering/genetics , Serogroup , Swine/virology
5.
Carcinogenesis ; 40(6): 707-714, 2019 07 06.
Article in English | MEDLINE | ID: mdl-31087000

ABSTRACT

Since its discovery, the E3 ubiquitin ligase E6-associated protein (E6AP) has been studied extensively in two pathological contexts: infection by the human papillomavirus (HPV), and the neurodevelopmental disorder, Angelman syndrome. Vital biological links between E6AP and other viruses, namely hepatitis C virus and encephalomyocarditis virus, have been recently uncovered. Critically, oncogenic E6AP activities have been demonstrated to contribute to cancers of both viral and non-viral origins. HPV-associated cancers serve as the primary example of E6AP involvement in cancers driven by viruses. Studies over the past few years have exposed a role for E6AP in non-viral-related cancers. This has been demonstrated in B-cell lymphoma and prostate cancers, where oncogenic E6AP functions drive these cancers by acting on key tumour suppressors. In this review we discuss the role of E6AP in viral infection, viral propagation and viral-related cancer. We discuss processes affected by oncogenic E6AP, which promote cancers of viral and non-viral aetiology. Overall, recent findings support the role of oncogenic E6AP in disrupting key cellular processes, including tumour suppression and the immune response. E6AP is consequently emerging as an attractive therapeutic target for a number of specific cancers.


Subject(s)
Neoplasms/physiopathology , Neoplasms/virology , Papillomavirus Infections/physiopathology , Ubiquitin-Protein Ligases/physiology , Carcinogenesis , Encephalomyocarditis virus/pathogenicity , Hepacivirus/pathogenicity , Host-Pathogen Interactions , Humans , Papillomaviridae/pathogenicity
6.
PLoS One ; 14(1): e0210971, 2019.
Article in English | MEDLINE | ID: mdl-30673749

ABSTRACT

Galectin-3 is a ß-galactoside-binding lectin which is important in cell proliferation and apoptotic regulation. Recently, serum galectin-3 has been shown to have prognostic value as a biomarker in heart failure. Encephalomyocarditis virus (EMCV) can cause severe myocarditis, congestive heart failure and dilated cardiomyopathy as well as encephalitis in various animals including mice. The pathophysiological role of galectin-3 in acute myocarditis following viral infection is not fully understood. The goal of this study is to determine the cardiac localization and the time-course of galectin-3 expression in heart failure after viral inoculation with EMCV. At 12, 24, 48, 96 hours, 7 and 10 days after intraperitoneal EMCV inoculation, animals were examined histologically and analyzed for the expression of galectin-3 and Iba1. Galectin-3 was up-regulated in degenerated fibrotic lesions of cardiac tissues 96 hours after viral inoculation and were followed by myocardial fibrosis. At the same time, Iba1 positive macrophages were observed within the inflammatory sites. A time-course correlation between the number of galectin-3 positive cells and the cardiac area of degenerated fibrotic lesions was detected-serum galectin-3 increased at 96 hours and correlated well with the number of cardiac galectin-3 positive cells. Our results indicate that galectin-3 expression may be a useful biomarker of cardiac fibrotic degeneration in acute myocarditis following viral infection. In addition, measuring serum galectin-3 levels might be an early diagnostic method for detecting cardiac degeneration in acute myocarditis.


Subject(s)
Cardiovirus Infections/blood , Cardiovirus Infections/metabolism , Encephalomyocarditis virus , Galectin 3/blood , Galectin 3/metabolism , Myocarditis/blood , Myocarditis/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/metabolism , Cardiovirus Infections/pathology , Disease Models, Animal , Encephalomyocarditis virus/pathogenicity , Fibrosis , Immunohistochemistry , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Prognosis , Sarcoglycans/deficiency , Sarcoglycans/genetics
7.
Virus Res ; 244: 164-172, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29113825

ABSTRACT

In 2007, numerous hamadryas baboons (Papio hamadryas) died suddenly in an aviary of a primate institute in Sochi, Russia, in the absence of prior clinical signs. Necropsies were suggestive of encephalomyocarditis virus infection, but RT-PCR assays with commonly used primers were negative. Here we report the histopathological results obtained during necropsies and the isolation and genomic characterization of a divergent strain of encephalomyocarditis virus 1 (EMCV-1) from heart tissue of one of the succumbed hamadryas baboons. Phylogenetic analysis indicates that the isolated virus belongs to the newly proposed EMCV-1 lineage G, which clusters alongside lineage C ("Mengo virus"). This study is the first report describing a lineage G strain of EMCV-1 as the etiological agent of a lethal disease outbreak among captive nonhuman primates in Europe.


Subject(s)
Cardiovirus Infections/epidemiology , Disease Outbreaks , Encephalomyocarditis virus/genetics , Genome, Viral , Papio hamadryas/virology , RNA, Viral/genetics , Amino Acid Sequence , Animals , Animals, Zoo , Autopsy , Cardiovirus Infections/mortality , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , DNA, Complementary/chemistry , DNA, Complementary/genetics , Encephalomyocarditis virus/classification , Encephalomyocarditis virus/isolation & purification , Encephalomyocarditis virus/pathogenicity , Heart/virology , Phylogeny , Russia/epidemiology , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid
8.
Virol J ; 14(1): 45, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28259172

ABSTRACT

BACKGROUD: Encephalomyocarditis virus (EMCV) has been discovered on pig farms worldwide and can cause myocarditis in piglets and reproductive failure in sows. However, little is known about the host transcriptional responses to infection and host-pathogen interactions. METHODS: In this study, transcription profiling was performed by Illumina RNA-Sequencing (RNA-seq) to identify EMCV induced differentially expressed genes in BHK-21 cells at serial time points (12, 24, and 30 h post infection (hpi)), using mock infected cells as control. RESULTS: We identified 237, 241, and 207 differentially expressed genes (DEGs) respectively, majority of which were up-regulated. A large number of DEGs clustered into host defense, cellular signaling and metabolism categories. Moreover, short time series expression analysis revealed that 12 hpi was an important time point for expression change, indicating host virus resistance. CONCLUSIONS: This RNA-seq analysis provides the first data for understanding the network of virus host interactions under EMCV infection in vitro, and for identifying host components which involved in the virus infection course.


Subject(s)
Encephalomyocarditis virus/immunology , Encephalomyocarditis virus/pathogenicity , Epithelial Cells/virology , Gene Expression Profiling , Host-Pathogen Interactions , Animals , Cell Line , Cricetinae , Sequence Analysis, RNA , Time Factors
9.
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
10.
J Gen Virol ; 97(9): 2280-2290, 2016 09.
Article in English | MEDLINE | ID: mdl-27392429

ABSTRACT

We successfully constructed a full-length cDNA infectious clone of the encephalomyocarditis virus (EMCV) HB10 strain and obtained a partially attenuated rEMCV-C9 virus with a shorter poly(C) tract. Our results showed that the length of the EMCV-HB10 poly(C) tract was related to the pathogenicity of the EMCV-HB10 strain in vivo. Using pEMCV-C9 as the backbone, we constructed the novel viral vector pC9-MCS-∆2A by inserting a cDNA fragment containing a 127 amino acid deletion in the 2A protein, a primary cleavage cassette, a FLAG tag and a multiple cloning site (MCS) at the junction of VP1 and ∆2A. Additionally, the enhanced green fluorescent protein (egfp) gene was cloned into the MCS of pC9-MCS-∆2A to test its capacity to express foreign proteins. Insertion of the egfp gene did not affect viral replication, and a decrease in EGFP expression was observed within five serial passages. Furthermore, we found that rC9-EGFP-∆2A was avirulent in vivo, induced neutralizing antibody production and conferred protective immune responses against lethal challenge with EMCV in mice. Taken together, our results demonstrated that we had constructed an attenuated live vector based on an EMCV-HB10 strain with two modified critical virulence factors (the poly(C) tract and 2A protein) that could be used as a candidate live vaccine and a potential live viral vector for foreign antigen delivery.


Subject(s)
Drug Carriers , Encephalomyocarditis virus/genetics , Genetic Engineering/methods , Genetic Vectors , Molecular Biology/methods , Animals , DNA, Complementary/genetics , DNA, Viral/genetics , Encephalomyocarditis virus/pathogenicity , Encephalomyocarditis virus/physiology , Genomic Instability , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Serial Passage , Technology, Pharmaceutical/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virus Replication
11.
Acta Virol ; 59(3): 300-4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26435154

ABSTRACT

Encephalomyocarditis virus (EMCV) can cause acute myocarditis in young pigs or reproductive failure in sows. The BD2 strain was isolated from the suspected piglets with EMCV in China. In order to establish an experimental animal model of EMCV, eight-weeks-old male BALB/c mice were intraperitonealy inoculated with 0.1 ml of 4×10(5) TCID50 suspension of the EMCV. Infected mice demonstrated hind limb paralysis, and movement disorder. The mortality rate of the infected group was 100% during the one-week observation period. The viral load in the brain of challenged mice gradually increased, with a peak level being 6.53 log CCID50/0.1 ml 5 days post infection. The pathological injury in infected mice was presented as neuronal necrosis. Brown positive staining could be detected in the cytoplasm of cerebral neurons. These results indicate that the porcine EMCV isolated from China could replicate in brain tissues and induce acute encephalitis in BALB/c mice.


Subject(s)
Cardiovirus Infections/etiology , Encephalomyocarditis virus/pathogenicity , Animals , Brain/virology , Male , Mice , Mice, Inbred BALB C , Viral Load
12.
J Virol ; 87(17): 9511-22, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23785203

ABSTRACT

In response to stress, cells induce ribonucleoprotein aggregates, termed stress granules (SGs). SGs are transient loci containing translation-stalled mRNA, which is eventually degraded or recycled for translation. Infection of some viruses, including influenza A virus with a deletion of nonstructural protein 1 (IAVΔNS1), induces SG-like protein aggregates. Previously, we showed that IAVΔNS1-induced SGs are required for efficient induction of type I interferon (IFN). Here, we investigated SG formation by different viruses using green fluorescent protein (GFP)-tagged Ras-Gap SH3 domain binding protein 1 (GFP-G3BP1) as an SG probe. HeLa cells stably expressing GFP-G3BP1 were infected with different viruses, and GFP fluorescence was monitored live with time-lapse microscopy. SG formations by different viruses was classified into 4 different patterns: no SG formation, stable SG formation, transient SG formation, and alternate SG formation. We focused on encephalomyocarditis virus (EMCV) infection, which exhibited transient SG formation. We found that EMCV disrupts SGs by cleavage of G3BP1 at late stages of infection (>8 h) through a mechanism similar to that used by poliovirus. Expression of a G3BP1 mutant that is resistant to the cleavage conferred persistent formation of SGs as well as an enhanced induction of IFN and other cytokines at late stages of infection. Additionally, knockdown of endogenous G3BP1 blocked SG formation with an attenuated induction of IFN and potentiated viral replication. Taken together, our findings suggest a critical role of SGs as an antiviral platform and shed light on one of the mechanisms by which a virus interferes with host stress and subsequent antiviral responses.


Subject(s)
Encephalomyocarditis virus/immunology , Encephalomyocarditis virus/pathogenicity , Ribonucleoproteins/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytokines/genetics , DNA Helicases , DNA Viruses/pathogenicity , Encephalomyocarditis virus/physiology , Gene Expression , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Interferons/genetics , Mutation , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , RNA Viruses/pathogenicity , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/immunology , Stress, Physiological , Virus Replication
13.
Virology ; 443(1): 177-85, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23711384

ABSTRACT

Encephalomyocarditis virus (EMCV), a Cardiovirus, initiates its polyprotein with a short 67 amino acid Leader (L) sequence. The protein acts as a unique pathogenicity factor, with anti-host activities which include the triggering of nuclear pore complex hyperphosphorylation and direct binding inhibition of the active cellular transport protein, Ran GTPase. Chemical modifications and protein mutagenesis now map the Ran binding domain to the L hinge-linker region, and in particular, to amino acids 35-40. Large deletions affecting this region were shown previously to diminish Ran binding. New point mutations, especially K35Q, D37A and W40A, preserve the intact L structure, abolish Ran binding and are deficient for nucleoporin (Nup) hyperphosphorylation. Ran itself morphs through multiple configurations, but reacts most effectively with L when in the GDP format, preferably with an empty nucleotide binding pocket. Therefore, L:Ran binding, mediated by the linker-hinge, is a required step in L-induced nuclear transport inhibition.


Subject(s)
Encephalomyocarditis virus/physiology , Host-Pathogen Interactions , Polyproteins/metabolism , Protein Interaction Mapping , Viral Proteins/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus , Amino Acid Substitution , Encephalomyocarditis virus/pathogenicity , HeLa Cells , Humans , Models, Molecular , Mutant Proteins/metabolism , Point Mutation , Protein Binding , Protein Conformation
14.
Can J Vet Res ; 76(2): 157-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-23024459

ABSTRACT

Encephalomyocarditis virus (EMCV) can infect many host species and cause acute myocarditis and sudden death in preweaned piglets. In this study, an EMCV strain (NJ08) was isolated from newborn pigs with clinical signs on a pig farm in mideastern China. It was identified by indirect immunofluorescence assay and reverse-transcription polymerase chain reaction. Experiments showed that the isolate could cause severe clinical symptoms and pathological changes in mice but no obvious clinical and pathological changes in commercial piglets. Complete genomic sequencing showed that the NJ08 strain was 78.3% to 100% identical with other isolates in regions coding for various proteins. Phylogenetic analysis showed that the NJ08 isolate belonged to subgroup Ia. This study confirmed that an EMCV isolate from pigs could be fatal to mice and provided new epidemiologic data on EMCV in China.


Subject(s)
Cardiovirus Infections/veterinary , Encephalomyocarditis virus/isolation & purification , Swine Diseases/virology , Animals , Animals, Newborn , Base Sequence , Biological Assay/veterinary , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cardiovirus Infections/virology , China , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/pathogenicity , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction/veterinary , RNA, Viral/chemistry , RNA, Viral/genetics , Sequence Alignment , Swine
15.
PLoS Pathog ; 8(7): e1002783, 2012.
Article in English | MEDLINE | ID: mdl-22792064

ABSTRACT

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Subject(s)
Antiviral Agents/pharmacology , Nitriles/pharmacology , Norovirus/drug effects , Pyridines/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/metabolism , Unfolded Protein Response/drug effects , Animals , Caliciviridae Infections/drug therapy , Caliciviridae Infections/virology , Cell Line , Cell Line, Tumor , Cyanoacrylates , DNA-Binding Proteins/metabolism , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/pathogenicity , Enzyme Inhibitors/pharmacology , Humans , La Crosse virus/drug effects , La Crosse virus/pathogenicity , Macrophages/virology , Membrane Proteins/metabolism , Mice , Norovirus/physiology , Norwalk virus/drug effects , Norwalk virus/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering , Regulatory Factor X Transcription Factors , Sindbis Virus/drug effects , Sindbis Virus/pathogenicity , Transcription Factors/metabolism , Ubiquitin/metabolism , Ubiquitin Thiolesterase/genetics , Virus Replication/drug effects , X-Box Binding Protein 1
16.
Virulence ; 3(4): 351-67, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22722247

ABSTRACT

The encephalomyocarditis virus (EMCV) is a small non-enveloped single-strand RNA virus, the causative agent of not only myocarditis and encephalitis, but also neurological diseases, reproductive disorders and diabetes in many mammalian species. EMCV pathogenesis appears to be viral strain- and host-specific, and a better understanding of EMCV virulence factors is increasingly required. Indeed, EMCV is often used as a model for diabetes and viral myocarditis, and is also widely used in immunology as a double-stranded RNA stimulus in the study of Toll-like as well as cytosolic receptors. However, EMCV virulence and properties have often been neglected. Moreover, EMCV is able to infect humans albeit with a low morbidity. Progress on xenografts, such as pig heart transplantation in humans, has raised safety concerns that need to be explored. In this review we will highlight the biology of EMCV and all known and potential virulence factors.


Subject(s)
Encephalomyocarditis virus/physiology , Encephalomyocarditis virus/pathogenicity , Host-Pathogen Interactions , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Cardiovirus Infections/veterinary , Cardiovirus Infections/virology , Humans , Mammals , Models, Biological , Virulence
17.
PLoS Pathog ; 8(5): e1002712, 2012.
Article in English | MEDLINE | ID: mdl-22615570

ABSTRACT

Interferon protects mice from vesicular stomatitis virus (VSV) infection and pathogenesis; however, it is not known which of the numerous interferon-stimulated genes (ISG) mediate the antiviral effect. A prominent family of ISGs is the interferon-induced with tetratricopeptide repeats (Ifit) genes comprising three members in mice, Ifit1/ISG56, Ifit2/ISG54 and Ifit3/ISG49. Intranasal infection with a low dose of VSV is not lethal to wild-type mice and all three Ifit genes are induced in the central nervous system of the infected mice. We tested their potential contributions to the observed protection of wild-type mice from VSV pathogenesis, by taking advantage of the newly generated knockout mice lacking either Ifit2 or Ifit1. We observed that in Ifit2 knockout (Ifit2(-/-)) mice, intranasal VSV infection was uniformly lethal and death was preceded by neurological signs, such as ataxia and hind limb paralysis. In contrast, wild-type and Ifit1(-/-) mice were highly protected and survived without developing such disease. However, when VSV was injected intracranially, virus replication and survival were not significantly different between wild-type and Ifit2(-/-) mice. When administered intranasally, VSV entered the central nervous system through the olfactory bulbs, where it replicated equivalently in wild-type and Ifit2(-/-) mice and induced interferon-ß. However, as the infection spread to other regions of the brain, VSV titers rose several hundred folds higher in Ifit2(-/-) mice as compared to wild-type mice. This was not caused by a broadened cell tropism in the brains of Ifit2(-/-) mice, where VSV still replicated selectively in neurons. Surprisingly, this advantage for VSV replication in the brains of Ifit2(-/-) mice was not observed in other organs, such as lung and liver. Pathogenesis by another neurotropic RNA virus, encephalomyocarditis virus, was not enhanced in the brains of Ifit2(-/-) mice. Our study provides a clear demonstration of tissue-, virus- and ISG-specific antiviral action of interferon.


Subject(s)
Brain/virology , Proteins/metabolism , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/pathogenicity , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Encephalomyocarditis virus/immunology , Encephalomyocarditis virus/pathogenicity , Female , Interferon-beta/metabolism , Liver/virology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteins/genetics , RNA-Binding Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Vesicular Stomatitis/pathology , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/immunology , Virus Replication
18.
Sci Rep ; 2: 367, 2012.
Article in English | MEDLINE | ID: mdl-22509465

ABSTRACT

Secondary bacterial infection in humans is one of the pathological conditions requiring clinical attention. In this study, we examined the effect of lipopolysaccharide (LPS) on encephalomyocarditis virus (EMCV) infected mice. All mice inoculated with EMCV at 5 days before LPS challenge died within 24 h. LPS-induced TNF-α mRNA expression was significantly increased in the brain and heart at 5 days after EMCV infection. CD11b(+)/TLR4(+) cell population in the heart was remarkably elevated at 5 days after EMCV infection, and sorted CD11b(+) cells at 5 days after EMCV infection produced a large amount of TNF-α on LPS stimulation in vivo and in vitro. In conclusion, we found that the infiltration of CD11b(+) cells into infected organs is involved in the subsequent LPS-induced lethal shock in viral encephalomyocarditis. This new experimental model can help define the mechanism by which secondary bacterial infection causes a lethal shock in viral encephalomyocarditis.


Subject(s)
Cardiovirus Infections/complications , Encephalomyocarditis virus/pathogenicity , Lipopolysaccharides/toxicity , Animals , Base Sequence , CD11b Antigen/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/metabolism , Cardiovirus Infections/mortality , DNA Primers , Flow Cytometry , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/biosynthesis
19.
Arch Virol ; 157(1): 43-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21989795

ABSTRACT

Encephalomyocarditis virus (EMCV) can infect a wide range of vertebrate species including swine and non-human primates, but few data are available for humans. We therefore wanted to gain further insight into the mechanisms involved in EMCV infection of human cells. For this purpose, we analyzed the permissiveness of primary human cardiomyocytes towards two strains of EMCV; a pig myocardial strain (B279/95) and a rat strain (1086C). In this study, we show that both strains productively infect primary human cardiomyocytes and induce complete cytolysis. Binding and infection inhibition experiments indicated that attachment and infection are independent of sialic acid and heparan sulfate for B279/95 and dependent for 1086C. Sequence comparison between the two strains and three-dimensional analysis of the capsid revealed that six of the seven variable residues are surface-exposed, suggesting a role for these amino acids in binding. Moreover, analysis of variants isolated from the 1086C strain revealed the importance of lysine 231 of VP1 in the attachment of EMCV to cell-surface sialic acid residues. Together, these results show a potential for EMCV strains to use at least two different binding possibilities to initiate infection and provide new insights into the mechanisms involved in primary human cell recognition by EMCV.


Subject(s)
Cardiovirus Infections/virology , Encephalomyocarditis virus/physiology , Encephalomyocarditis virus/pathogenicity , Myocytes, Cardiac/virology , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cells, Cultured , Encephalomyocarditis virus/classification , Encephalomyocarditis virus/genetics , Genetic Variation , Humans , Molecular Sequence Data , Virulence
20.
J Virol ; 85(20): 10741-54, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849462

ABSTRACT

The encephalomyocarditis virus (EMCV), a Picornaviridae virus, has a wide host spectrum and can cause various diseases. EMCV virulence factors, however, are as yet ill defined. Here, we demonstrate that the EMCV 2A protein is essential for the pathogenesis of EMCV. Infection of mice with the B279/95 strain of EMCV resulted in acute fatal disease, while the clone C9, derived by serial in vitro passage of the B279/95 strain, was avirulent. C9 harbored a large deletion in the gene encoding the 2A protein. This deletion was incorporated into the cDNA of a pathogenic EMCV1.26 strain. The new virus, EMCV1.26Δ2A, was capable of replicating in vitro, albeit more slowly than EMCV1.26. Only mice inoculated with EMCV1.26 triggered death within a few days. Mice infected with EMCV1.26Δ2A did not exhibit clinical signs, and histopathological analyses showed no damage in the central nervous system, unlike EMCV1.26-infected mice. In vitro, EMCV1.26Δ2A presented a defect in viral particle release correlating with prolonged cell viability. Unlike EMCV1.26, which induced cytopathic cell death, EMCV1.26Δ2A induced apoptosis via caspase 3 activation. This strongly suggests that the 2A protein is required for inhibition of apoptosis during EMCV infection. All together, our data indicate that the EMCV 2A protein is important for the virus in counteracting host defenses, since Δ2A viruses were no longer pathogenic and were unable to inhibit apoptosis in vitro.


Subject(s)
Apoptosis , Encephalomyocarditis virus/pathogenicity , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , Cell Line , Cell Survival , Central Nervous System/pathology , Cricetinae , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mutant Proteins/genetics , Mutant Proteins/metabolism , Rodent Diseases/pathology , Rodent Diseases/virology , Sequence Deletion , Survival Analysis , Viral Proteins/genetics , Virulence Factors/genetics , Virus Release , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...