1
|
Tamás V, Righi C, Mészáros I, D'Errico F, Olasz F, Casciari C, Zádori Z, Magyar T, Petrini S, Feliziani F. Involvement of the MGF 110-11L Gene in the African Swine Fever Replication and Virulence. Vaccines (Basel) 2023; 11:vaccines11040846. [PMID: 37112759 PMCID: PMC10145817 DOI: 10.3390/vaccines11040846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
African swine fever (ASF) is a highly lethal hemorrhagic viral disease that causes extensive economic and animal welfare losses in the Eurasian pig (Sus scrofa) population. To date, no effective and safe vaccines have been marketed against ASF. A starting point for vaccine development is using naturally occurring attenuated strains as a vaccine base. Here, we aimed to remove the multigene family (MGF) 110 gene of unknown function from the Lv17/WB/Rie1 genome to improve the usability of the virus as a live-attenuated vaccine, reducing unwanted side effects. The MGF 110-11L gene was deleted using the CRISPR/Cas9 method, and the safety and efficacy of the virus were tested in pigs after isolation. The vaccine candidates administered at high doses showed reduced pathogenicity compared to the parental strain and induced immunity in vaccinated animals, although several mild clinical signs were observed. Although Lv17/WB/Rie1/d110-11L cannot be used as a vaccine in its current form, it was encouraging that the undesirable side effects of Lv17/WB/Rie1 at high doses can be reduced by additional mutations without a significant reduction in its protective capacity.
Collapse
Affiliation(s)
- Vivien Tamás
- Institute for Veterinary Medical Research, Hungária krt. 21, 1143 Budapest, Hungary
| | - Cecilia Righi
- Istituto Zooprofilattico Sperimentale Umbria-Marche "Togo Rosati", Via Gaetano Salvemini, 1, 06126 Perugia, Italy
| | - István Mészáros
- Institute for Veterinary Medical Research, Hungária krt. 21, 1143 Budapest, Hungary
| | - Federica D'Errico
- Istituto Zooprofilattico Sperimentale Umbria-Marche "Togo Rosati", Via Gaetano Salvemini, 1, 06126 Perugia, Italy
| | - Ferenc Olasz
- Institute for Veterinary Medical Research, Hungária krt. 21, 1143 Budapest, Hungary
| | - Cristina Casciari
- Istituto Zooprofilattico Sperimentale Umbria-Marche "Togo Rosati", Via Gaetano Salvemini, 1, 06126 Perugia, Italy
| | - Zoltán Zádori
- Institute for Veterinary Medical Research, Hungária krt. 21, 1143 Budapest, Hungary
| | - Tibor Magyar
- Institute for Veterinary Medical Research, Hungária krt. 21, 1143 Budapest, Hungary
| | - Stefano Petrini
- Istituto Zooprofilattico Sperimentale Umbria-Marche "Togo Rosati", Via Gaetano Salvemini, 1, 06126 Perugia, Italy
| | - Francesco Feliziani
- Istituto Zooprofilattico Sperimentale Umbria-Marche "Togo Rosati", Via Gaetano Salvemini, 1, 06126 Perugia, Italy
| |
Collapse
|
2
|
Penza V, Maroun JW, Nace RA, Schulze AJ, Russell SJ. Polycytidine tract deletion from microRNA-detargeted oncolytic Mengovirus optimizes the therapeutic index in a murine multiple myeloma model. Mol Ther Oncolytics 2023; 28:15-30. [PMID: 36619293 PMCID: PMC9800256 DOI: 10.1016/j.omto.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Mengovirus is an oncolytic picornavirus whose broad host range allows for testing in immunocompetent cancer models. Two pathogenicity-ablating approaches, polycytidine (polyC) tract truncation and microRNA (miRNA) targets insertion, eliminated the risk of encephalomyocarditis. To investigate whether a polyC truncated, miRNA-detargeted oncolytic Mengovirus might be boosted, we partially or fully rebuilt the polyC tract into the 5' noncoding region (NCR) of polyC-deleted (MC0) oncolytic constructs (NC) carrying miRNA target (miRT) insertions to eliminate cardiac/muscular (miR-133b and miR-208a) and neuronal (miR-124) tropisms. PolyC-reconstituted viruses (MC24-NC and MC37-NC) replicated in vitro and showed the expected tropism restrictions, but reduced cytotoxicity and miRT deletions were frequently observed. In the MPC-11 immune competent mouse plasmacytoma model, both intratumoral and systemic administration of MC0-NC led to faster tumor responses than MC24-NC or MC37-NC, with combined durable complete response rates of 75%, 0.5%, and 30%, respectively. Secondary viremia was higher following MC0-NC versus MC24-NC or MC37-NC therapy. Sequence analysis of virus progeny from treated mice revealed a high prevalence of miRT sequences loss among MC24- and MC37- viral genomes, but not in MC0-NC. Overall, MC0-NC was capable of stably retaining miRT sites and provided a more effective treatment and is therefore our lead Mengovirus candidate for clinical translation.
Collapse
Affiliation(s)
- Velia Penza
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55902, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Justin W. Maroun
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN 55902, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Autumn J. Schulze
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Stephen J. Russell
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN 55902, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Insights from structural studies of the Cardiovirus 2A protein. Biosci Rep 2022; 42:230648. [PMID: 35022657 PMCID: PMC8777194 DOI: 10.1042/bsr20210406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/24/2022] Open
Abstract
Cardioviruses are single-stranded RNA viruses of the family Picornaviridae. In addition to being the first example of internal ribosome entry site (IRES) utilization, cardioviruses also employ a series of alternative translation strategies, such as Stop-Go translation and programmed ribosome frameshifting. Here, we focus on cardiovirus 2A protein, which is not only a primary virulence factor, but also exerts crucial regulatory functions during translation, including activation of viral ribosome frameshifting and inhibition of host cap-dependent translation. Only recently, biochemical and structural studies have allowed us to close the gaps in our knowledge of how cardiovirus 2A is able to act in diverse translation-related processes as a novel RNA-binding protein. This review will summarize these findings, which ultimately may lead to the discovery of other RNA-mediated gene expression strategies across a broad range of RNA viruses.
Collapse
|
4
|
Tan YL, Tan KSW, Chu JJH, Chow VT. Combination Treatment With Remdesivir and Ivermectin Exerts Highly Synergistic and Potent Antiviral Activity Against Murine Coronavirus Infection. Front Cell Infect Microbiol 2021; 11:700502. [PMID: 34395311 PMCID: PMC8362885 DOI: 10.3389/fcimb.2021.700502] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/13/2021] [Indexed: 12/16/2022] Open
Abstract
The recent COVID-19 pandemic has highlighted the urgency to develop effective antiviral therapies against the disease. Murine hepatitis virus (MHV) is a coronavirus that infects mice and shares some sequence identity to SARS-CoV-2. Both viruses belong to the Betacoronavirus genus, and MHV thus serves as a useful and safe surrogate model for SARS-CoV-2 infections. Clinical trials have indicated that remdesivir is a potentially promising antiviral drug against COVID-19. Using an in vitro model of MHV infection of RAW264.7 macrophages, the safety and efficacy of monotherapy of remdesivir, chloroquine, ivermectin, and doxycycline were investigated. Of the four drugs tested, remdesivir monotherapy exerted the strongest inhibition of live virus and viral RNA replication of about 2-log10 and 1-log10, respectively (at 6 µM). Ivermectin treatment showed the highest selectivity index. Combination drug therapy was also evaluated using remdesivir (6 µM) together with chloroquine (15 µM), ivermectin (2 µM) or doxycycline (15 µM) - above their IC50 values and at high macrophage cell viability of over 95%. The combination of remdesivir and ivermectin exhibited highly potent synergism by achieving significant reductions of about 7-log10 of live virus and 2.5-log10 of viral RNA in infected macrophages. This combination also resulted in the lowest cytokine levels of IL-6, TNF-α, and leukemia inhibitory factor. The next best synergistic combination was remdesivir with doxycycline, which decreased levels of live virus by ~3-log10 and viral RNA by ~1.5-log10. These results warrant further studies to explore the mechanisms of action of the combination therapy, as well as future in vivo experiments and clinical trials for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yu Ling Tan
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Kevin S. W. Tan
- Healthy Longevity Translational Research Program, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Vincent T. Chow
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
5
|
Comparative analysis, distribution, and characterization of microsatellites in Orf virus genome. Sci Rep 2020; 10:13852. [PMID: 32807836 PMCID: PMC7431841 DOI: 10.1038/s41598-020-70634-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/01/2020] [Indexed: 11/09/2022] Open
Abstract
Genome-wide in-silico identification of microsatellites or simple sequence repeats (SSRs) in the Orf virus (ORFV), the causative agent of contagious ecthyma has been carried out to investigate the type, distribution and its potential role in the genome evolution. We have investigated eleven ORFV strains, which resulted in the presence of 1,036-1,181 microsatellites per strain. The further screening revealed the presence of 83-107 compound SSRs (cSSRs) per genome. Our analysis indicates the dinucleotide (76.9%) repeats to be the most abundant, followed by trinucleotide (17.7%), mononucleotide (4.9%), tetranucleotide (0.4%) and hexanucleotide (0.2%) repeats. The Relative Abundance (RA) and Relative Density (RD) of these SSRs varied between 7.6-8.4 and 53.0-59.5 bp/kb, respectively. While in the case of cSSRs, the RA and RD ranged from 0.6-0.8 and 12.1-17.0 bp/kb, respectively. Regression analysis of all parameters like the incident of SSRs, RA, and RD significantly correlated with the GC content. But in a case of genome size, except incident SSRs, all other parameters were non-significantly correlated. Nearly all cSSRs were composed of two microsatellites, which showed no biasedness to a particular motif. Motif duplication pattern, such as, (C)-x-(C), (TG)-x-(TG), (AT)-x-(AT), (TC)- x-(TC) and self-complementary motifs, such as (GC)-x-(CG), (TC)-x-(AG), (GT)-x-(CA) and (TC)-x-(AG) were observed in the cSSRs. Finally, in-silico polymorphism was assessed, followed by in-vitro validation using PCR analysis and sequencing. The thirteen polymorphic SSR markers developed in this study were further characterized by mapping with the sequence present in the database. The results of the present study indicate that these SSRs could be a useful tool for identification, analysis of genetic diversity, and understanding the evolutionary status of the virus.
Collapse
|
6
|
Van Borm S, Fu Q, Winand R, Vanneste K, Hakhverdyan M, Höper D, Vandenbussche F. Evaluation of a commercial exogenous internal process control for diagnostic RNA virus metagenomics from different animal clinical samples. J Virol Methods 2020; 283:113916. [PMID: 32574649 DOI: 10.1016/j.jviromet.2020.113916] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022]
Abstract
Metagenomic next generation sequencing (mNGS) is increasingly recognized as an important complementary tool to targeted human and animal infectious disease diagnostics. It is, however, sensitive to biases and errors that are currently not systematically evaluated by the implementation of quality controls (QC) for the diagnostic use of mNGS. We evaluated a commercial reagent (Mengovirus extraction control kit, CeraamTools, bioMérieux) as an exogenous internal control for mNGS. It validates the integrity of reagents and workflow, the efficient isolation of viral nucleic acids and the absence of inhibitors in individual samples (verified using a specific qRT-PCR). Moreover, it validates the efficient generation of viral sequence data in individual samples (verified by normalized mengoviral read counts in the metagenomic analysis). We show that when using a completely random metagenomics workflow: (1) Mengovirus RNA can be reproducibly detected in different animal sample types (swine feces and sera, wild bird cloacal swabs), except for tissue samples (swine lung); (2) the Mengovirus control kit does not contain other contaminating viruses that may affect metagenomic experiments (using a cutoff of minimum 1 Kraken classified read per million (RPM)); (3) the addition of 2.17 × 106Mengovirus copies/mL of sample does not affect the virome composition of pig fecal samples or wild bird cloacal swab samples; (4) Mengovirus Cq values (using as cutoff the upper limit of the 99 % confidence interval of Cq values for a given sample matrix) allow the identification of samples with poor viral RNA extraction or high inhibitor load; (5) Mengovirus normalized read counts (cutoff RPM > 1) allow the identification of samples where the viral sequences are outcompeted by host or bacterial target sequences in the random metagenomic workflow. The implementation of two QC testing points, a first one after RNA extraction (Mengoviral qRT-PCR) and a second one after metagenomic data analysis provide valuable information for the validation of individual samples and results. Their implementation in addition to external controls validating runs or experiments should be carefully considered for a given sample type and workflow.
Collapse
Affiliation(s)
- Steven Van Borm
- Department of Animal Infectious Diseases, Sciensano, Groeselenbergstraat 99, 1180, Brussels, Belgium.
| | - Qiang Fu
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Raf Winand
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsmanstraat 14, 1050, Brussels, Belgium
| | - Kevin Vanneste
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsmanstraat 14, 1050, Brussels, Belgium
| | | | - Dirk Höper
- FLI, Friedrich Löffler Institut, Südufer 10, 17493 Greifswald, Germany
| | - Frank Vandenbussche
- Department of Animal Infectious Diseases, Sciensano, Groeselenbergstraat 99, 1180, Brussels, Belgium
| |
Collapse
|
7
|
Stewart H, Olspert A, Butt BG, Firth AE. Propensity of a picornavirus polymerase to slip on potyvirus-derived transcriptional slippage sites. J Gen Virol 2018; 100:199-205. [PMID: 30507373 PMCID: PMC6591135 DOI: 10.1099/jgv.0.001189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The substitution rates of viral polymerases have been studied extensively. However less is known about the tendency of these enzymes to 'slip' during RNA synthesis to produce progeny RNAs with nucleotide insertions or deletions. We recently described the functional utilization of programmed polymerase slippage in the family Potyviridae. This slippage results in either an insertion or a substitution, depending on whether the RNA duplex realigns following the insertion. In this study we investigated whether this phenomenon is a conserved feature of superfamily I viral RdRps, by inserting a range of potyvirus-derived slip-prone sequences into a picornavirus, Theiler's murine encephalomyelitis virus (TMEV). Deep-sequencing analysis of viral transcripts indicates that the TMEV polymerase 'slips' at the sequences U6-7 and A6-7 to insert additional nucleotides. Such sequences are under-represented within picornaviral genomes, suggesting that slip-prone sequences create a fitness cost. Nonetheless, the TMEV insertional and substitutional spectrum differed from that previously determined for the potyvirus polymerase.
Collapse
Affiliation(s)
- Hazel Stewart
- 1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Allan Olspert
- 2School of Science, Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Benjamin G Butt
- 1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Andrew E Firth
- 1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Binding interactions between the encephalomyocarditis virus leader and protein 2A. J Virol 2014; 88:13503-9. [PMID: 25210192 DOI: 10.1128/jvi.02148-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
UNLABELLED The leader (L) and 2A proteins of cardioviruses are the primary antihost agents produced during infection. For encephalomyocarditis virus (EMCV), the prototype of the genus Cardiovirus, these proteins interact independently with key cellular partners to bring about inhibition of active nucleocytoplasmic trafficking and cap-dependent translation, respectively. L and 2A also bind each other and require this cooperation to achieve their effects during infection. Recombinant L and 2A interact with 1:1 stoichiometry at a KD (equilibrium dissociation constant) of 1.5 μM. The mapped contact domains include the amino-proximal third of 2A (first 50 amino acids) and the central hinge region of L. This contact partially overlaps the L segment that makes subsequent contact with Ran GTPase in the nucleus, and Ran can displace 2A from L. The equivalent proteins from Theiler's murine encephalomyelitis virus (TMEV; BeAn) and Saffold virus interact similarly in any subtype combination, with various affinities. The data suggest a mechanism whereby L takes advantage of the nuclear localization signal in the COOH region of 2A to enhance its trafficking to the nucleus. Once there, it exchanges partners in favor of Ran. This required cooperation during infection explains many observed codependent phenotypes of L and 2A mutations. IMPORTANCE Cardiovirus pathogenesis phenotypes vary dramatically, from asymptomatic, to mild gastrointestinal (GI) distress, to persistent demyelination and even encephalitic death. Leader and 2A are the primary viral determinants of pathogenesis, so understanding how these proteins cooperate to induce such a wide variety of outcomes for the host is of great important and interest to the field of virology, especially to those who use TMEV as a murine model for multiple sclerosis.
Collapse
|
9
|
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.
Collapse
Affiliation(s)
- Margot Carocci
- Microbiology Immunology Department, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
10
|
Bai J, Jiang K, Zhang G, Wang X, Li Y, Jiang P. Pathogenicity and molecular analysis of an encephalomyocarditis virus isolate from mideastern China. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2012; 76:157-160. [PMID: 23024459 PMCID: PMC3314439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 05/07/2011] [Indexed: 06/01/2023]
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.
Collapse
Affiliation(s)
| | | | | | | | | | - Ping Jiang
- Address all correspondence to Dr. Ping Jiang; telephone: 86 25 84395504; fax: 86 25 84396640; e-mail:
| |
Collapse
|
11
|
Rosenthal LA, Szakaly RJ, Amineva SP, Xing Y, Hill MR, Palmenberg AC, Gern JE, Sorkness RL. Lower respiratory tract infection induced by a genetically modified picornavirus in its natural murine host. PLoS One 2012; 7:e32061. [PMID: 22355409 PMCID: PMC3280220 DOI: 10.1371/journal.pone.0032061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 01/23/2012] [Indexed: 12/21/2022] Open
Abstract
Infections with the picornavirus, human rhinovirus (HRV), are a major cause of wheezing illnesses and asthma exacerbations. In developing a murine model of picornaviral airway infection, we noted the absence of murine rhinoviruses and that mice are not natural hosts for HRV. The picornavirus, mengovirus, induces lethal systemic infections in its natural murine hosts, but small genetic differences can profoundly affect picornaviral tropism and virulence. We demonstrate that inhalation of a genetically attenuated mengovirus, vMC0, induces lower respiratory tract infections in mice. After intranasal vMC0 inoculation, lung viral titers increased, peaking at 24 h postinoculation with viral shedding persisting for 5 days, whereas HRV-A01a lung viral titers decreased and were undetectable 24 h after intranasal inoculation. Inhalation of vMC0, but not vehicle or UV-inactivated vMC0, induced an acute respiratory illness, with body weight loss and lower airway inflammation, characterized by increased numbers of airway neutrophils and lymphocytes and elevated pulmonary expression of neutrophil chemoattractant CXCR2 ligands (CXCL1, CXCL2, CXCL5) and interleukin-17A. Mice inoculated with vMC0, compared with those inoculated with vehicle or UV-inactivated vMC0, exhibited increased pulmonary expression of interferon (IFN-α, IFN-β, IFN-λ), viral RNA sensors [toll-like receptor (TLR)3, TLR7, nucleotide-binding oligomerization domain containing 2 (NOD2)], and chemokines associated with HRV infection in humans (CXCL10, CCL2). Inhalation of vMC0, but not vehicle or UV-inactivated vMC0, was accompanied by increased airway fluid myeloperoxidase levels, an indicator of neutrophil activation, increased MUC5B gene expression, and lung edema, a sign of infection-related lung injury. Consistent with experimental HRV inoculations of nonallergic, nonasthmatic human subjects, there were no effects on airway hyperresponsiveness after inhalation of vMC0 by healthy mice. This novel murine model of picornaviral airway infection and inflammation should be useful for defining mechanisms of HRV pathogenesis in humans.
Collapse
Affiliation(s)
- Louis A Rosenthal
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Rosenthal LA, Amineva SP, Szakaly RJ, Lemanske RF, Gern JE, Sorkness RL. A rat model of picornavirus-induced airway infection and inflammation. Virol J 2009; 6:122. [PMID: 19671179 PMCID: PMC2790594 DOI: 10.1186/1743-422x-6-122] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 08/11/2009] [Indexed: 11/12/2022] Open
Abstract
Background Infection of the lower airways by rhinovirus, a member of the picornavirus family, is an important cause of wheezing illnesses in infants, and plays an important role in the pathogenesis of rhinovirus-induced asthma exacerbations. Given the absence of natural rhinovirus infections in rodents, we investigated whether an attenuated form of mengovirus, a picornavirus whose wild-type form causes systemic rather than respiratory infections in its natural rodent hosts, could induce airway infections in rats with inflammatory responses similar to those in human rhinovirus infections. Results After inoculation with 107 plaque-forming units of attenuated mengovirus through an inhalation route, infectious mengovirus was consistently recovered on days 1 and 3 postinoculation from left lung homogenates (median Log10 plaque-forming units = 6.0 and 4.8, respectively) and right lung bronchoalveolar lavage fluid (median Log10 plaque-forming units = 5.8 and 4.0, respectively). Insufflation of attenuated mengovirus, but not vehicle or UV-inactivated virus, into the lungs of BN rats caused significant increases (P < 0.05) in lower airway neutrophils and lymphocytes in the bronchoalveolar lavage fluid and patchy peribronchiolar, perivascular, and alveolar cellular infiltrates in lung tissue sections. In addition, infection with attenuated mengovirus significantly increased (P < 0.05) lower airway levels of neutrophil chemoattractant CXCR2 ligands [cytokine-induced neutrophil chemoattractant-1 (CINC-1; CXCL1) and macrophage inflammatory protein-2 (MIP-2; CXCL2)] and monocyte chemoattractant protein-1 (MCP-1; CCL2) in comparison to inoculation with vehicle or UV-inactivated virus. Conclusion Attenuated mengovirus caused a respiratory infection in rats with several days of viral shedding accompanied by a lower airway inflammatory response consisting of neutrophils and lymphocytes. These features suggest that mengovirus-induced airway infection in rodents could be a useful model to define mechanisms of rhinovirus-induced airway inflammation in humans.
Collapse
Affiliation(s)
- Louis A Rosenthal
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
In Canada, cardiovirus isolates related to Saffold virus were detected in nasopharyngeal aspirates from 3 children with respiratory symptoms. Polyprotein sequence of the Can112051-06 isolate had 91.2% aa identity with Saffold virus; however, EF and CD loops of the viral surface varied substantially.
Collapse
Affiliation(s)
- Yacine Abed
- Centre Hospitalier Universitaire de Québec, Quebec City, Canada
| | | |
Collapse
|
14
|
Stuhlmeier KM. Hyaluronan production in synoviocytes as a consequence of viral infections: HAS1 activation by Epstein-Barr virus and synthetic double- and single-stranded viral RNA analogs. J Biol Chem 2008; 283:16781-9. [PMID: 18400745 DOI: 10.1074/jbc.m801669200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
One of the hallmarks of arthritis is swollen joints containing unusually high quantities of hyaluronan. Intact hyaluronan molecules facilitate cell migration by acting as ligands for CD44. Hyaluronan degradation products, readily formed at sites of inflammation, also fuel inflammatory processes. Irrespective of whether viruses could be a cause of rheumatoid arthritis, there is clear evidence that links viral infections to this debilitating disease. For this study, live Epstein-Barr virus and a number of double- and single-stranded synthetic viral analogs were tested for their effectiveness as activators of hyaluronan (HA) synthesis. As shown herein, Epstein-Barr virus-treated fibroblast-like synoviocytes significantly increase HA production and release. Real time reverse transcription-PCR data show that HAS1 mRNA levels are significantly elevated in virus-treated cells, whereas mRNA levels for the genes HAS2 and HAS3 remain unchanged. As to the mechanism of virus-induced HAS1 transcription, data are presented that imply that among the double- and single-stranded polynucleotides tested, homopolymeric polycytidylic structures are the most potent inducers of HAS1 transcription and HA release, whereas homopolymeric polyinosinic acid is without effect. Analyses of virus-induced signal cascades, utilizing chemical inhibitors of MAPK and overexpressing mutated IKK and IkappaB, revealed that the MAPK p38 as well as the transcription factor NF-kappaB are essential for virus-induced activation of HAS1. The presented data implicate HAS1 as the culprit in unfettered HA release and point out targets in virus-induced signaling pathways that might allow for specific interventions in cases of unwanted and uncontrolled HA synthesis.
Collapse
Affiliation(s)
- Karl M Stuhlmeier
- Ludwig Boltzmann Institute for Rheumatology and Balneology, Kurbadstrasse 10, 1100 Vienna, Austria.
| |
Collapse
|
15
|
Zarudnaya MI, Stepanyugin AV, Potyahaylo AL, Hovorun DM. Detection of two different protonated forms of poly(C) by electrophoresis method. ACTA ACUST UNITED AC 2008. [DOI: 10.7124/bc.000792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- M. I. Zarudnaya
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - A. V. Stepanyugin
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - A. L. Potyahaylo
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - D. M. Hovorun
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| |
Collapse
|
16
|
Dyer KD, Schellens IMM, Bonville CA, Martin BV, Domachowske JB, Rosenberg HF. Efficient replication of pneumonia virus of mice (PVM) in a mouse macrophage cell line. Virol J 2007; 4:48. [PMID: 17547763 PMCID: PMC1891281 DOI: 10.1186/1743-422x-4-48] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 06/04/2007] [Indexed: 01/04/2023] Open
Abstract
Pneumonia virus of mice (PVM; family Paramyxoviridae, subfamily Pneumovirinae) is a natural respiratory pathogen of rodent species and an important new model for the study of severe viral bronchiolitis and pneumonia. However, despite high virus titers typically detected in infected mouse lung tissue in vivo, cell lines used routinely for virus propagation in vitro are not highly susceptible to PVM infection. We have evaluated several rodent and primate cell lines for susceptibility to PVM infection, and detected highest virus titers from infection of the mouse monocyte-macrophage RAW 264.7 cell line. Additionally, virus replication in RAW 264.7 cells induces the synthesis and secretion of proinflammatory cytokines relevant to respiratory virus disease, including tumor necrosis factor-alpha (TNF-alpha), interferon-beta (IFN-beta), macrophage inflammatory proteins 1alpha and 1beta (MIP-1alpha and MIP-1beta) and the functional homolog of human IL-8, mouse macrophage inflammatory peptide-2 (MIP-2). Identification and characterization of a rodent cell line that supports the replication of PVM and induces the synthesis of disease-related proinflammatory mediators will facilitate studies of molecular mechanisms of viral pathogenesis that will complement and expand on findings from mouse model systems.
Collapse
Affiliation(s)
- Kimberly D Dyer
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ingrid MM Schellens
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cynthia A Bonville
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Brittany V Martin
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Pharmacology, University of Colorado Health Sciences Center, Denver, Colorado, USA
| | - Joseph B Domachowske
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Helene F Rosenberg
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Building 10, Room 11C215, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
17
|
Jones MS, Lukashov VV, Ganac RD, Schnurr DP. Discovery of a novel human picornavirus in a stool sample from a pediatric patient presenting with fever of unknown origin. J Clin Microbiol 2007; 45:2144-50. [PMID: 17460053 PMCID: PMC1933019 DOI: 10.1128/jcm.00174-07] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fever of unknown origin (FUO) is a serious problem in the United States. An unidentified agent was cultured from the stool of an infant who presented with FUO. This virus showed growth in HFDK cells and suckling mice. Using DNase sequence-independent single-primer amplification, we identified several nucleotide sequences with a high homology to Theiler's murine encephalomyelitis virus. Nearly full-length viral genome sequencing and phylogenetic analysis demonstrate that this virus is a member of the Cardiovirus genus of the Picornaviridae family.
Collapse
Affiliation(s)
- Morris S Jones
- Clinical Investigation Facility, David Grant USAF Medical Center, 101 Bodin Circle, Travis AFB, CA 94535, USA.
| | | | | | | |
Collapse
|
18
|
Tracy S, Chapman NM, Drescher KM, Kono K, Tapprich W. Evolution of virulence in picornaviruses. Curr Top Microbiol Immunol 2006; 299:193-209. [PMID: 16568900 DOI: 10.1007/3-540-26397-7_7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The Picornaviridae encompass many positive-strand RNA viruses, all of which share a generally similar genome design and capsid structure, but which induce quite diverse diseases in humans and other animals. Picornavirus strains of the same serotype have been shown to express different virulence (or pathogenic) phenotypes when studied in animal models, demonstrating that key elements of pathogenesis reside in the viral genome. However, the genetics that determine the virulence phenotype of any picornavirus are poorly understood. Picornaviruses do not have virulence genes per se, but the design ofthe capsid andhow it interacts with the virus receptor expressed on the host cell surface, specific sequences within the nontranslated regions of the viral genome, as well as coding sequences that result in different protein sequences may all have a part in determining the virulence phenotype. Virulence may be better understood as a continuum from an apparent inability to induce disease to the ability to cause severe pathogenic changes. Ultimately, the ability of a picornavirus to induce disease depends upon viral genetics and how they are modulated by the host environment.
Collapse
Affiliation(s)
- S Tracy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-6495, USA.
| | | | | | | | | |
Collapse
|
19
|
Denis P, Liebig HD, Nowotny N, Billinis C, Papadopoulos O, O'Hara RS, Knowles NJ, Koenen F. Genetic variability of encephalomyocarditis virus (EMCV) isolates. Vet Microbiol 2006; 113:1-12. [PMID: 16406410 DOI: 10.1016/j.vetmic.2005.10.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2004] [Revised: 10/05/2005] [Accepted: 10/13/2005] [Indexed: 11/20/2022]
Abstract
In order to evaluate the variability of encephalomyocarditis virus (EMCV), field isolates originating from different European regions and inducing different clinical pictures in pigs have been molecularly characterised. The regions targeted were the poly(C) tract, a part of the 5'-UTR (360 nucleotides), the Leader gene (201 nucleotides), the complete capsid coding region (2502 nucleotides), the 2A gene (403 nucleotides), the end of the 3D polymerase gene (305 nucleotides) and the 3'-UTR (123 nucleotides). Analyses have also been performed on a virulent field isolate, which had been subjected to serial passages in vivo and in vitro resulting, in the case of the in vitro passaged virus, in attenuation, as demonstrated by animal experiments. The present study shows that different clinical pictures, such as acute fatal myocarditis or reproductive failure, may not only be caused by EMCV isolates which are genetically diverse but also by the same isolate. Thus no correlation could be demonstrated between genotype and clinical disease. However, the European isolate which showed the highest genetic divergence also gave rise to a more complex clinical picture. Despite EMCV having been isolated from cases of acute fatal myocarditis in pigs in certain areas of the world for many years, clinical disease, including a variety of clinical pictures and pathogenicity, has only been recognised in Europe since 1986 and thus it can be considered an emerging disease in this region. These findings, associated with the reported phenotype changes of the virus under environmental changes (passages), along with its wide distribution among vertebrate species (including higher primates), shows the validity of considering EMCV as a potential pathogen for recipients in xenotransplantation.
Collapse
Affiliation(s)
- P Denis
- Veterinary and Agrochemical Research Center, Groeselenberg 99, 1180 Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Moran JM, Moxley MA, Buller RML, Corbett JA. Encephalomyocarditis virus induces PKR-independent mitogen-activated protein kinase activation in macrophages. J Virol 2005; 79:10226-36. [PMID: 16051816 PMCID: PMC1182679 DOI: 10.1128/jvi.79.16.10226-10236.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this study, we provide evidence that the double-stranded RNA-dependent protein kinase (PKR) is not required for virus-induced expression of inducible nitric oxide synthase (iNOS) or the activation of specific signaling pathways in macrophages. The infection of RAW264.7 cells with encephalomyocarditis virus (EMCV) induces iNOS expression and nitric oxide production, which are unaffected by a dominant-negative mutant of PKR. EMCV infection also activates the mitogen-activated protein kinase, cyclic AMP response element binding protein, and nuclear factor kappaB (NF-kappaB) signaling cascades at 15 to 30 min postinfection in PKR+/+ and PKR-/- macrophages. Activation of these signaling cascades does not temporally correlate with PKR activity or the accumulation of EMCV RNA, suggesting that an interaction between a structural component of the virion and the cell surface may activate macrophages. Consistent with this hypothesis, empty EMCV capsids induced comparable levels of iNOS expression, nitrite production, and activation of these signaling cascades to those induced by intact virions. These findings support the hypothesis that virion-host cell interactions are primary mediators of the PKR-independent activation of signaling pathways that participate in the macrophage antiviral response of inflammatory gene expression.
Collapse
Affiliation(s)
- Jason M Moran
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1402 South Grand Boulevard, Saint Louis, MO 63104, USA
| | | | | | | |
Collapse
|
21
|
Karalyan ZA, Jaghatspanyan NG, Gasparyan MH, Hakobyan LA, Abroyan LO, Ter-Pogossyan ZR, Kamalyan LA, Karalova EM. Comparison of impact of EMCV replication on the nuclear apparatus of NIH 3T3 and HEp-2 cells. Cell Biol Int 2005; 29:586-92. [PMID: 15996489 DOI: 10.1016/j.cellbi.2005.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Revised: 02/06/2005] [Accepted: 02/24/2005] [Indexed: 10/25/2022]
Abstract
We have investigated differences between the actions of encephalomyocarditis virus (EMCV) on cytometric indices in cultured NIH 3T3 and HEp-2 cells, which are characterized by different levels of transformation. HEp-2 cells surviving 48 h after EMCV infection showed lower nuclear ploidy, reduced nuclear area, fewer nucleoli and a higher percentage of euploid cells. There was a significant increase of nucleolar/nuclear DNA 6-24 h after EMCV infection. However, EMCV had markedly different effects on NIH 3T3 cells: there was a consistent increase in population ploidy, but the average number of nucleoli and the number of euploid cells in the population remained constant. The nucleolar/nuclear DNA ratio was almost unchanged. These different viral effects might be explained by the contrasting levels of differentiation of the cultured cell lines. The number of nucleoli does not depend on the amount of nuclear DNA in either viral-infected or intact cells but on the euploidy-to-aneuploidy ratio. The ratio of the sums of the nucleolar perimeters to the nuclear perimeter increases linearly with the number of nucleoli per nucleus in both intact and virus-infected cells. In both cell lines, the amount of DNA per nucleolus decreases as the number of nucleoli increases.
Collapse
Affiliation(s)
- Z A Karalyan
- Laboratory of Virology, Cancer Research Center, Yerevan, Armenia.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
LaRue R, Myers S, Brewer L, Shaw DP, Brown C, Seal BS, Njenga MK. A wild-type porcine encephalomyocarditis virus containing a short poly(C) tract is pathogenic to mice, pigs, and cynomolgus macaques. J Virol 2003; 77:9136-46. [PMID: 12915530 PMCID: PMC187386 DOI: 10.1128/jvi.77.17.9136-9146.2003] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2003] [Accepted: 06/03/2003] [Indexed: 12/22/2022] Open
Abstract
Previous studies using wild-type Encephalomyocarditis virus (EMCV) and Mengo virus, which have long poly(C) tracts (61 to 146 C's) at the 5' nontranslated region of the genome, and variants of these viruses genetically engineered to truncate or substitute the poly(C) tracts have produced conflicting data on the role of the poly(C) tract in the virulence of these viruses. Analysis of the nucleotide sequence of an EMCV strain isolated from an aborted swine fetus (EMCV 30/87) revealed that the virus had a poly(C) tract that was 7- to 10-fold shorter than the poly(C) tracts of other EMCV strains and 4-fold shorter than that of Mengo virus. Subsequently, we investigated the virulence and pathogenesis of this naturally occurring short-poly(C)-tract-containing virus in rodents, pigs, and nonhuman primates. Infection of C57BL/6 mice, pigs, and cynomolgus macaques resulted in similar EMCV 30/87 pathogenesis, with the heart and brain as the primary sites of infections in all three animals, but with different disease phenotypes. Sixteen percent of EMCV 30/87-infected pigs developed acute fatal cardiac failure, whereas the rest of the pigs were overtly asymptomatic for as long as 90 days postinfection (p.i.), despite extensive myocardial and central nervous system (CNS) pathological changes. In contrast, mice infected with >/==" BORDER="0">4 PFU of EMCV 30/87 developed acute encephalitis that resulted in the death of all animals (n = 25) between days 2 and 7 p.i. EMCV 30/87-infected macaques remained overtly asymptomatic for 45 days, despite extensive myocardial and CNS pathological changes and viral persistence in more than 50% of the animals. The short poly(C) tract in EMCV 30/87 (CUC(5)UC(8)) was comparable to that of strain 2887A/91 (C(10)UCUC(3)UC(10)), another recent porcine isolate.
Collapse
Affiliation(s)
- Rebecca LaRue
- Department of Veterinary Pathobiology, University of Minnesota, St. Paul, Minnesota 55108, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Aminev AG, Amineva SP, Palmenberg AC. Encephalomyocarditis viral protein 2A localizes to nucleoli and inhibits cap-dependent mRNA translation. Virus Res 2003; 95:45-57. [PMID: 12921995 DOI: 10.1016/s0168-1702(03)00162-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Panels of monoclonal antibodies were raised against viral non-structural proteins of encephalomyocarditis virus (EMCV) and used to probe infected cells in laser confocal microscopy experiments and Western analyses. Surprisingly, all Mengovirus and EMCV-infected cells showed strong targeting of protein 2A, 3B(VPg), 3C(pro), and 3D(pol) signals to cellular nuclei, in particular to nucleoli, from the earliest times of infection. Viral capsid proteins (1AB, 1C, and 1D) and other non-structural proteins (2B, 2C, and 3A) did not target nuclei and remained cytoplasmic throughout the infection. The cardioviral 2A protein (subject of this article) has a novel 143 amino acid sequence, terminating in a 19 amino acid COOH-terminal processing cassette (PCC) that participates in autocatalytic, co-translational primary cleavage of the viral polyprotein. The remainder of the 2A protein shares only limited similarity with other viral or cellular sequences, except for a short motif (KRvRPFRLP) near PCC resembling the nuclear localization signals (NLS) common to many yeast ribosomal proteins. Deletions within the EMCV 2A protein that impinge on this region have been reported to diminish the ability of virus to inhibit cap-dependent translation of cellular mRNAs. We have now observed that these same deletions prevented nuclear localization. Cellular expression of 2A protein from RNA transcripts or cDNAs confirmed that it does not require other viral proteins or activities for nuclear transport; even when expressed as a single protein, 2A protein effectively shuts off translation from capped reporter mRNAs. Within infected, transfected, or DNA vector-transformed cells, the 2A protein was always found in close association with the nucleolar ribosomal chaperone protein B23, which may help the traffic 2A into nucleoli like a surrogate ribosomal protein, by virtue of the putative nucleolar localization signal (NoLS). The data are consistent with a novel mechanism for virus-induced host protein shut off in cardioviruses, whereby 2A helps to upregulate the synthesis of new and modified ribosomes that have an inherent preference for internal ribosomal entry site (IRES)-dependent viral genome translation over cap-dependent host mRNA translation.
Collapse
Affiliation(s)
- Aleksey G Aminev
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | | | | |
Collapse
|
24
|
Lyons AJ, Robertson HD. Detection of tRNA-like structure through RNase P cleavage of viral internal ribosome entry site RNAs near the AUG start triplet. J Biol Chem 2003; 278:26844-50. [PMID: 12746454 DOI: 10.1074/jbc.m304052200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The 9600-base RNA genome of hepatitis C virus (HCV) has an internal ribosome entry site (IRES) in its first 370 bases, including the AUG start triplet at bases 342-344. Structural elements of this and other IRES domains substitute for a 5' terminal cap structure in protein synthesis. Recent work (Nadal, A., Martell, M., Lytle, J. R., Lyons, A. J., Robertson, H. D., Cabot, B., Esteban, J. I., Esteban, R., Guardia, J., and Gomez, J. (2002) J. Biol. Chem. 277, 30606-30613) has demonstrated that the host pre-tRNA processing enzyme, RNase P, can cleave the HCV RNA genome at a site in the IRES near the AUG initiator triplet. Although this step is unlikely to be part of the HCV life cycle, such a reaction could indicate the presence of a tRNA-like structure in this IRES. Because susceptibility to cleavage by mammalian RNase P is a strong indicator of tRNA-like structure, we have conducted the studies reported here to test whether such tRNA mimicry is unique to HCV or is a general property of IRES structure. We have assayed IRES domains of several viral RNA genomes: two pestiviruses related to HCV, classical swine fever virus and bovine viral diarrhea virus; and two unrelated viruses, encephalomyocarditis virus and cricket paralysis virus. We have found similarly placed RNase P cleavage sites in these IRESs. Thus a tRNA-like domain could be a general structural feature of IRESs, the first IRES structure to be identified with a functional correlate. Such tRNA-like features could be recognized by pre-existing ribosomal tRNA-binding sites as part of the IRES initiation cycle.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites/genetics
- Classical Swine Fever Virus/genetics
- Classical Swine Fever Virus/metabolism
- Codon, Initiator/chemistry
- Diarrhea Viruses, Bovine Viral/genetics
- Diarrhea Viruses, Bovine Viral/metabolism
- Endoribonucleases/metabolism
- Hepacivirus/genetics
- Hepacivirus/metabolism
- Humans
- Nucleic Acid Conformation
- RNA, Catalytic/metabolism
- RNA, Transfer/chemistry
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- RNA, Viral/chemistry
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Ribonuclease P
Collapse
Affiliation(s)
- Alita J Lyons
- Department of Biochemistry, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | |
Collapse
|
25
|
Abstract
To define structural elements critical for RNA replication in human parechovirus 1 (HPeV1), a replicon with chloramphenicol acetyltransferase as a reporter gene and an infectious virus cDNA clone have been used. It was observed that there are cis-acting signals required for HPeV1 replication located within the 5'-terminal 112 nucleotides of the genome and that these include two terminal stem-loops, SL-A and SL-B, together with a pseudoknot element. Significant disruption of any of these structures impaired both RNA replication and virus growth. In view of the similarity in terminal structures to several picornaviruses, such as cardioviruses and hepatoviruses, the insights generated in this work are of wider significance for understanding picornavirus replication.
Collapse
Affiliation(s)
- Abdolrahman S Nateri
- Department of Biological Sciences, University of Essex, Colchester CO4 3SQ, United Kingdom
| | | | | |
Collapse
|
26
|
Zarudnaya MI, Samijlenko SP, Potyahaylo AL, Hovorun DM. Structural transitions in polycytidylic acid: proton buffer capacity data. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2002; 21:125-37. [PMID: 11991140 DOI: 10.1081/ncn-120003177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The pH-dependences of proton buffer capacity of poly(C) were computed on the basis of the literature data. In these curves there were observed four peaks: two narrow and two wide ones. The first narrow peak reflects the process of cooperative formation of double helices, which is induced by protonation of the N3 atom of nucleotide bases. The first wide peak is assigned to noncooperative process of poly(C) double helices protonation at the N3 nitrogen atom. It is proposed that the second wide peak corresponds to noncooperative protonation of the neutral cytosine bases at the oxygen atom. This reaction causes cooperative dissociation of the poly(C) double helices. The second narrow peak reflects the dissociation process.
Collapse
Affiliation(s)
- Margarita I Zarudnaya
- Molecular Biophysics Department, Institute of Molecular Biology and Genetics, Ukrainian National Academy of Sciences, Kyiv
| | | | | | | |
Collapse
|
27
|
Neal ZC, Harms JS, Hill MR, Splitter GA. Encephalomyocarditis and Mengo viruses productively infect murine T-lymphocyte cell lines but not fresh ex vivo derived T lymphocytes. Viral Immunol 2002; 15:155-63. [PMID: 11952137 DOI: 10.1089/088282402317340305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Encephalomyocarditis virus (EMCV) and Mengo virus are highly virulent murine cardioviruses that are found in abundant quantities in the spleen and lymph nodes after infection. T lymphocytes are pivotal mediators of humoral and cellular immunity against cardioviral challenge, and are highly suspect candidates of EMCV and Mengo virus infection. We found T lymphocyte-like cell lines CTLL-2, EL-4, LY1+2/9, and LBRM33 were susceptible to productive viral infection and exhibited cytopathology after infection with virulent EMCV-R or attenuated Mengo virus strains vMC0 and vMC24. Flow cytometric analysis demonstrated progressive intracellular accumulation of viral proteins, such as the replication-dependent 3D viral polymerase, in EL-4 cells during infection. Conversely, freshly isolated and mitogen-stimulated CD4+ and CD8+ T cells were resistant to productive infection with these viruses, exhibiting no viral-induced cytopathic effects or intracellular presence of viral proteins. These data indicate that although T-lymphocyte-like tumor cell lines are highly susceptible to viral infection and cytopathic effects, primary/freshly isolated T cells are resistant to infection by EMCV-R or Mengo virus.
Collapse
Affiliation(s)
- Zane C Neal
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison, 53706-1581, USA
| | | | | | | |
Collapse
|
28
|
Kassimi LB, Boutrouille A, Gonzague M, Mbanda AL, Cruciere C. Nucleotide sequence and construction of an infectious cDNA clone of an EMCV strain isolated from aborted swine fetus. Virus Res 2002; 83:71-87. [PMID: 11864742 DOI: 10.1016/s0168-1702(01)00425-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A full-length cDNA clone of an Encephalomyocarditis virus (EMCV) strain (2887A) isolated from aborted swine fetus was constructed and sequenced. Sequence comparison showed more than 99% nucleotide and amino acid sequence identity with two other EMCV strains, EMCV-PV21 and -R. However, the 2887A genomic sequence showed only about 84% nucleotide identity and 96% amino acid identity with EMCV-B, -D and -PV2 variants. RNA synthesized by in vitro transcription of this cDNA clone was infectious upon transfection of BHK21 cells, as shown by cytopathic effects and identification by neutralization test, and by propagation of the virus released into the culture media. The transcript RNA led to the production of infectious particles despite the presence of two nongenomic nucleotide residues at the 5' end, the short poly(C) tract (C(10)TCTC(3)TC(10)), the short poly(A) tail (7A), and the presence of six nongenomic nucleotides at the 3' end. The rescued virus was also found to be highly pathogenic for mice by intra-peritoneal inoculation producing a fatal disease indistinguishable from that of wild-type virus. An important finding concerning the molecular basis of infectivity was that the in vitro synthesized EMCV RNA transcript is infectious, although it contains a very short poly(A). The availability of the infectious cDNA clone of the reproductive failure strain of EMCV should prove to be useful for studying the molecular basis of the pathogenicity of EMCV in pig.
Collapse
Affiliation(s)
- Labib Bakkali Kassimi
- Agence Française de Sécurité Sanitaire des Aliments, AFSSA-Alfort, 22 rue Pierre Curie BP 67, 94703 Maisons-Alfort Cedex, France.
| | | | | | | | | |
Collapse
|
29
|
Dodd DA, Giddings TH, Kirkegaard K. Poliovirus 3A protein limits interleukin-6 (IL-6), IL-8, and beta interferon secretion during viral infection. J Virol 2001; 75:8158-65. [PMID: 11483761 PMCID: PMC115060 DOI: 10.1128/jvi.75.17.8158-8165.2001] [Citation(s) in RCA: 132] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
During viral infections, the host secretory pathway is crucial for both innate and acquired immune responses. For example, the export of most proinflammatory and antiviral cytokines, which recruit lymphocytes and initiate antiviral defenses, requires traffic through the host secretory pathway. To investigate potential effects of the known inhibition of cellular protein secretion during poliovirus infection on pathogenesis, cytokine secretion from cells infected with wild-type virus and with 3A-2, a mutant virus carrying an insertion in viral protein 3A which renders the virus defective in the inhibition of protein secretion, was tested. We show here that cells infected with 3A-2 mutant virus secrete greater amounts of cytokines interleukin-6 (IL-6), IL-8, and beta interferon than cells infected with wild-type poliovirus. Increased cytokine secretion from the mutant-infected cells can be attributed to the reduced inhibition of host protein secretion, because no significant differences between 3A-2- and wild-type-infected cells were observed in the inhibition of viral growth, host cell translation, or the ability of wild-type- or 3A-2-infected cells to support the transcriptional induction of beta interferon mRNA. We surmise that the wild-type function of 3A in inhibiting ER-to-Golgi traffic is not required for viral replication in tissue culture but, by altering the amount of secreted cytokines, could have substantial effects on pathogenesis within an infected host. The global inhibition of protein secretion by poliovirus may reflect a general mechanism by which pathogens that do not require a functional protein secretory apparatus can reduce the native immune response and inflammation associated with infection.
Collapse
Affiliation(s)
- D A Dodd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94309, USA
| | | | | |
Collapse
|