Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Sci Rep ; 9(1): 7216, 2019 05 10.
Article in English | MEDLINE | ID: mdl-31076606

ABSTRACT

The sensing of viral nucleic acids by the innate immune system activates a potent antiviral response in the infected cell, a key component of which is the expression of genes encoding type I interferons (IFNs). Many viruses counteract this response by blocking the activation of host nucleic acid sensors. The evolutionarily conserved influenza A virus (IAV) protein PA-X has been implicated in suppressing the host response to infection, including the expression of type I IFNs. Here, we characterise this further using a PA-X-deficient virus of the mouse-adapted PR8 strain to study activation of the innate immune response in a mouse model of the early response to viral infection. We show that levels of Ifna4 and Ifnb1 mRNAs in the lungs of infected mice were elevated in the absence of PA-X and that this was completely dependent on MAVS. This therefore suggests a role for PA-X in preventing the accumulation of early type I IFN mRNAs in the lung during IAV infection.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Influenza A virus/physiology , Interferon Type I/metabolism , Repressor Proteins/genetics , Viral Nonstructural Proteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Immunity, Innate , Influenza A virus/metabolism , Interferon Type I/genetics , Lung/metabolism , Lung/virology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , RNA, Messenger/metabolism , Repressor Proteins/deficiency , Repressor Proteins/metabolism , Signal Transduction , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/metabolism
2.
Sci Rep ; 7(1): 329, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28336944

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. KSHV establishes a life-long infection in its host and alternates between a latent and lytic infection state. During lytic infection, lytic-related genes are expressed in a temporal manner and categorized as immediate early, early, and late gene transcripts. ORF34 is an early-late gene that interacts with several viral transcription-associated factors, however its physiological importance remains poorly understood. Here, we investigated the role of ORF34 during KSHV infection by generating ORF34-deficient KSHV, using a bacterial artificial chromosome system. Our results reveal that ORF34-deficient KSHV exhibited significantly attenuated late gene expression and viral production but did not affect viral DNA replication. ORF34 interacted with transcription factors ORF18, ORF24, ORF31, and ORF66, and a novel ORF34-interaction partner, ORF23. The C-terminal region of ORF34 was important for interaction with ORF24 and viral production. Our data support a model, in which ORF34 serves as a hub for recruiting a viral transcription complex to ORF24 to promote late viral gene expression.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Transcription, Genetic , Viral Nonstructural Proteins/metabolism , Animals , Chlorocebus aethiops , Gene Knockout Techniques , Herpesvirus 8, Human/growth & development , Protein Interaction Mapping , Vero Cells , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/genetics , Virus Replication
3.
Med Microbiol Immunol ; 205(4): 381-95, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27289459

ABSTRACT

PA-X is a novel discovered accessory protein encoded by the PA mRNA. Our previous study demonstrated that PA-X decreases the virulence of a highly pathogenic H5N1 strain A/Chicken/Jiangsu/k0402/2010 in mice. However, the underlying mechanism of virulence attenuation associated with PA-X is still unknown. In this study, we compared two PA-X-deficient mutant viruses and the parental virus in terms of induction of pathology and manipulation of host response in the mouse lung, stimulation of cell death and PA nuclear accumulation. We first found that down-regulated PA-X expression markedly aggravated the acute lung injury of the infected mice early on day 1 post-infection (p.i.). We then determined that loss of PA-X expression induced higher levels of cytokines, chemokines and complement-derived peptides (C3a and C5a) in the lung, especially at early time point's p.i. In addition, in vitro assays showed that the PA-X-deficient viruses enhanced cell death and increased expression of reactive oxygen species (ROS) in mammalian cells. Moreover, we also found that PA nuclear accumulation of the PA-X-null viruses accelerated in MDCK cells. These results demonstrate that PA-X decreases the level of complement components, ROS, cell death and inflammatory response, which may together contribute to the alleviated lung injury and the attenuation of the virulence of H5N1 virus in mice.


Subject(s)
Acute Lung Injury/pathology , Acute Lung Injury/virology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Repressor Proteins/immunology , Viral Nonstructural Proteins/immunology , Animals , Cell Death , Complement System Proteins/analysis , Cytokines/analysis , Disease Models, Animal , Dogs , Female , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Lung/pathology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Reactive Oxygen Species/analysis , Repressor Proteins/deficiency , Viral Nonstructural Proteins/deficiency
4.
J Virol ; 90(5): 2616-27, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26699638

ABSTRACT

UNLABELLED: Oropouche virus (OROV) is a midge-borne human pathogen with a geographic distribution in South America. OROV was first isolated in 1955, and since then, it has been known to cause recurring outbreaks of a dengue-like illness in the Amazonian regions of Brazil. OROV, however, remains one of the most poorly understood emerging viral zoonoses. Here we describe the successful recovery of infectious OROV entirely from cDNA copies of its genome and generation of OROV mutant viruses lacking either the NSm or the NSs coding region. Characterization of the recombinant viruses carried out in vitro demonstrated that the NSs protein of OROV is an interferon (IFN) antagonist as in other NSs-encoding bunyaviruses. Additionally, we demonstrate the importance of the nine C-terminal amino acids of OROV NSs in IFN antagonistic activity. OROV was also found to be sensitive to IFN-α when cells were pretreated; however, the virus was still capable of replicating at doses as high as 10,000 U/ml of IFN-α, in contrast to the family prototype BUNV. We found that OROV lacking the NSm protein displayed characteristics similar to those of the wild-type virus, suggesting that the NSm protein is dispensable for virus replication in the mammalian and mosquito cell lines that were tested. IMPORTANCE: Oropouche virus (OROV) is a public health threat in Central and South America, where it causes periodic outbreaks of dengue-like illness. In Brazil, OROV is the second most frequent cause of arboviral febrile illness after dengue virus, and with the current rates of urban expansion, more cases of this emerging viral zoonosis could occur. To better understand the molecular biology of OROV, we have successfully rescued the virus along with mutants. We have established that the C terminus of the NSs protein is important in interferon antagonism and that the NSm protein is dispensable for virus replication in cell culture. The tools described in this paper are important in terms of understanding this important yet neglected human pathogen.


Subject(s)
Host-Pathogen Interactions , Orthobunyavirus/genetics , Orthobunyavirus/physiology , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line , Humans , Interferons/antagonists & inhibitors , Orthobunyavirus/immunology , Reverse Genetics
5.
J Virol ; 89(20): 10273-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26223635

ABSTRACT

UNLABELLED: The NS1 protein of influenza virus has multiple functions and is a determinant of virulence. Influenza viruses with NS1 deletions (DelNS1 influenza viruses) are a useful tool for studying virus replication and can serve as effective live attenuated vaccines, but deletion of NS1 severely diminishes virus replication, hampering functional studies and vaccine production. We found that WSN-DelNS1 viruses passaged in cells consistently adapted to gain an A14U substitution in the 3' noncoding region of the M segment of viral RNA (vRNA) which restored replicative ability. DelNS1-M-A14U viruses cannot inhibit interferon expression in virus infected-cells, providing an essential model for studying virus replication in the absence of the NS1 protein. Characterization of DelNS1-M-A14U virus showed that the lack of NS1 has no apparent effect on expression of other viral proteins, with the exception of M mRNAs. Expression of the M transcripts, M1, M2, mRNA3, and mRNA4, is regulated by alternative splicing. The A14U substitution changes the splicing donor site consensus sequence of mRNA3, altering expression of M transcripts, with M2 expression significantly increased and mRNA3 markedly suppressed in DelNS1-M-A14U, but not DelNS1-M-WT, virus-infected cells. Further analysis revealed that the A14U substitution also affects promoter function during replication of the viral genome. The M-A14U mutation increases M vRNA synthesis in DelNS1 virus infection and enhances alternative splicing of M2 mRNA in the absence of other viral proteins. The findings demonstrate that NS1 is directly involved in influenza virus replication through modulation of alternative splicing of M transcripts and provide strategic information important to construction of vaccine strains with NS1 deletions. IMPORTANCE: Nonstructural protein (NS1) of influenza virus has multiple functions. Besides its role in antagonizing host antiviral activity, NS1 is also believed to be involved in regulating virus replication, but mechanistic details are not clear. The NS1 protein is a virulence determinant which inhibits both innate and adaptive immunity and live attenuated viruses with NS1 deletions show promise as effective vaccines. However, deletion of NS1 causes severe attenuation of virus replication during infection, impeding functional studies and vaccine development. We characterized a replication-competent DelNS1 virus which carries an A14U substitution in the 3' noncoding region of the vRNA M segment. We found that M-A14U mutation supports virus replication through modulation of alternative splicing of mRNAs transcribed from the M segment. Our findings give insight into the role of NS1 in influenza virus replication and provide an approach for constructing replication-competent strains with NS1 deletions for use in functional and vaccine studies.


Subject(s)
Alternative Splicing , Genome, Viral , Influenza A virus/genetics , RNA, Viral/genetics , Viral Nonstructural Proteins/genetics , Virus Replication/genetics , 3' Untranslated Regions , Animals , Base Sequence , Chlorocebus aethiops , Dogs , HEK293 Cells , Humans , Influenza A virus/metabolism , Madin Darby Canine Kidney Cells , Molecular Sequence Data , Point Mutation , RNA Splice Sites , RNA, Viral/metabolism , Sequence Deletion , Vero Cells , Viral Nonstructural Proteins/deficiency
6.
ACS Nano ; 9(3): 2696-703, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25560916

ABSTRACT

The polyvinylpyrrolidone (PVP)-coated spherically clustered porous gold-silver alloy nanoparticle (PVP-SPAN) was prepared by low temperature mediated, partially inhibited galvanic replacement reaction followed by silver etching process. The prepared porous nanostructures exhibited excellent photothermal conversion efficiency under irradiation of near-infrared light (NIR) and allowed a high payload of both doxorubicin (Dox) and thiolated dye-labeled oligonucleotide, DNAzyme (FDz). Especially, PVP-SPAN provided 10 times higher loading capacity for oligonucleotide than conventional hollow nanoshells due to increased pore diameter and surface-to-volume ratio. We demonstrated highly efficient chemo-thermo-gene multitherapy based on codelivery of Dox and FDz with NIR-mediated photothermal therapeutic effect using a model system of hepatitis C virus infected human liver cells (Huh7 human hepatocarcinoma cell line containing hepatitis C virus NS3 gene replicon) compared to conventional hollow nanoshells.


Subject(s)
Alloys/chemistry , Drug Carriers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Nanotechnology/methods , Silver/chemistry , Cell Line, Tumor , Combined Modality Therapy , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Liberation , Hepacivirus/drug effects , Hepacivirus/physiology , Hepacivirus/radiation effects , Humans , Hydrogen Peroxide/chemistry , Infrared Rays , Models, Molecular , Molecular Conformation , Porosity , Povidone/chemistry , Temperature , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/genetics
7.
Vaccine ; 33(6): 812-8, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25510389

ABSTRACT

Bluetongue is a disease of ruminants caused by the bluetongue virus (BTV). Bluetongue outbreaks can be controlled by vaccination, however, currently available vaccines have several drawbacks. Further, there are at least 26 BTV serotypes, with low cross protection. A next-generation vaccine based on live-attenuated BTV without expression of non-structural proteins NS3/NS3a, named Disabled Infectious Single Animal (DISA) vaccine, was recently developed for serotype 8 by exchange of the serotype determining outer capsid protein VP2. DISA vaccines are replicating vaccines but do not cause detectable viremia, and induce serotype specific protection. Here, we exchanged VP2 of laboratory strain BTV1 for VP2 of European serotypes 2, 4, 8 and 9 using reverse genetics, without observing large effects on virus growth. Exchange of VP2 from serotype 16 and 25 was however not possible. Therefore, chimeric VP2 proteins of BTV1 containing possible immunogenic regions of these serotypes were studied. BTV1, expressing 1/16 chimeric VP2 proteins was functional in virus replication in vitro and contained neutralizing epitopes of both serotype 1 and 16. For serotype 25 this approach failed. We combined VP2 exchange with the NS3/NS3a negative phenotype in BTV1 as previously described for serotype 8 DISA vaccine. DISA vaccine with 1/16 chimeric VP2 containing amino acid region 249-398 of serotype 16 raised antibodies in sheep neutralizing both BTV1 and BTV16. This suggests that DISA vaccine could be protective for both parental serotypes present in chimeric VP2. We here demonstrate the application of the BT DISA vaccine platform for several serotypes and further extend the application for serotypes that are unsuccessful in single VP2 exchange.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/immunology , Bluetongue virus/immunology , Bluetongue/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Antigens, Viral/genetics , Bluetongue/immunology , Bluetongue/virology , Bluetongue virus/classification , Bluetongue virus/drug effects , Bluetongue virus/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Cross Protection , Epitopes/genetics , Epitopes/immunology , Female , Gene Expression , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Reverse Genetics , Serogroup , Sheep , Vaccination , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Replication
8.
J Virol ; 88(17): 10157-64, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965464

ABSTRACT

UNLABELLED: Viral proteins often display several functions which require multiple assays to dissect their genetic basis. Here, we describe a systematic approach to screen for loss-of-function mutations that confer a fitness disadvantage under a specified growth condition. Our methodology was achieved by genetically monitoring a mutant library under two growth conditions, with and without interferon, by deep sequencing. We employed a molecular tagging technique to distinguish true mutations from sequencing error. This approach enabled us to identify mutations that were negatively selected against, in addition to those that were positively selected for. Using this technique, we identified loss-of-function mutations in the influenza A virus NS segment that were sensitive to type I interferon in a high-throughput fashion. Mechanistic characterization further showed that a single substitution, D92Y, resulted in the inability of NS to inhibit RIG-I ubiquitination. The approach described in this study can be applied under any specified condition for any virus that can be genetically manipulated. IMPORTANCE: Traditional genetics focuses on a single genotype-phenotype relationship, whereas high-throughput genetics permits phenotypic characterization of numerous mutants in parallel. High-throughput genetics often involves monitoring of a mutant library with deep sequencing. However, deep sequencing suffers from a high error rate (∼0.1 to 1%), which is usually higher than the occurrence frequency for individual point mutations within a mutant library. Therefore, only mutations that confer a fitness advantage can be identified with confidence due to an enrichment in the occurrence frequency. In contrast, it is impossible to identify deleterious mutations using most next-generation sequencing techniques. In this study, we have applied a molecular tagging technique to distinguish true mutations from sequencing errors. It enabled us to identify mutations that underwent negative selection, in addition to mutations that experienced positive selection. This study provides a proof of concept by screening for loss-of-function mutations on the influenza A virus NS segment that are involved in its anti-interferon activity.


Subject(s)
Influenza A virus/immunology , Influenza A virus/physiology , Interferon Type I/antagonists & inhibitors , Mutation , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/metabolism , High-Throughput Nucleotide Sequencing , Influenza A virus/genetics , Influenza A virus/growth & development , Molecular Biology/methods , RNA, Viral/genetics , Virology/methods
9.
Vaccine ; 31(52): 6194-200, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24183981

ABSTRACT

BACKGROUND: The non-structural protein NS1 of the influenza virus counteracts the interferon-mediated immune response of the host. We investigated the safety and immunogenicity of a trivalent formulation containing influenza H1N1, H3N2 and B strains lacking NS1 (delNS1-trivalent). METHODS: Healthy adult study participants who were seronegative for at least one strain present in the vaccine formulation were randomized to receive a single intranasal dose of delNS1-trivalent vaccine at 7.0log10 TCID50/subject (n=39) or placebo (n=41). RESULTS: Intranasal vaccination with the live replication-deficient delNS1-trivalent vaccine was well tolerated with no treatment-related serious adverse events. The most common adverse events identified, i.e. headache, oropharyngeal pain and rhinitis-like symptoms, were mainly mild and transient and distributed similarly in the treatment and placebo groups. Significant vaccine-specific immune responses were induced. Pre-existing low antibody titers or seronegativity for the corresponding vaccine strain yielded better response rates. CONCLUSIONS: We show that vaccination with a replication-deficient trivalent influenza vaccine containing H1N1, H3N2 and B strains lacking NS1 is safe and induces significant levels of antibodies (ClinicalTrials.gov identifier NCT01369862).


Subject(s)
Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Viral Nonstructural Proteins/deficiency , Administration, Intranasal , Adolescent , Adult , Antibodies, Viral/blood , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Healthy Volunteers , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/genetics , Influenza B virus/immunology , Influenza Vaccines/administration & dosage , Male , Placebos/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
10.
Vopr Virusol ; 58(3): 28-31, 2013.
Article in Russian | MEDLINE | ID: mdl-24006630

ABSTRACT

In the present work, the immunoadjuvant properties of the influenza deltaNS1 vaccine virus after intranasal administration in combination with recombinant GBS polypeptides was tested in mice. According to our data, co-administration of recombinant GBS polypeptides and influenza deltaNS1 vaccine resulted in the increase in the immunogenicity and protective efficacy of bacterial proteins. Combined vaccination with the GBS polypeptides and influenza deltaNS1 vaccine has a potential to be used not only for prophylaxis infections caused by SGB, but also for prevention of the bacterial complications of influenza.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Viral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antibodies, Viral/blood , Cross Protection , Female , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Peptides/genetics , Peptides/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/genetics , Vaccination , Vaccines, Synthetic , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/genetics
11.
J Virol ; 87(4): 2320-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23236071

ABSTRACT

Hepatitis C virus NS5A has three structural domains, is required for RNA replication and virion assembly, and exists in hypo- and hyperphosphorylated forms. Accumulated data suggest that phosphorylation is involved in modulating NS5A functions. We performed a mutational analysis of highly conserved serine residues in the linker region between domains I and II of genotype 2a JFH1 NS5A. As with genotype 1b Con1 NS5A, we found that specific serine residues were important for efficient hyperphosphorylation of JFH1 NS5A. However, in contrast with Con1 replicons, we observed a positive correlation between hyperphosphorylation and JFH1 replicon replication. We previously demonstrated trans-complementation of a hyperphosphorylation-deficient, replication-defective JFH1 replicon. Our results suggested that the defective NS5A encoded by this replicon, while lacking one NS5A function, was capable of performing a separate replication function. In this report, we examined an additional set of replication-defective NS5A mutations in trans-complementation assays. While some behaved similarly to the S232I replicon, others displayed a unique trans-complementation phenotype, suggesting that NS5A trans-complementation can occur by two distinct modes. Moreover, we were able, for the first time, to demonstrate intragenic complementation of replication-defective NS5A alleles. Our results identified three complementation groups: group A, comprising mutations within NS5A domain I; group B, comprising mutations affecting serine residues important for hyperphosphorylation and a subset of the domain I mutations; and group C, comprising a single mutation within the C-terminal region of domain II. We postulate that these complementation groups define three distinct and genetically separable functions of NS5A in RNA replication.


Subject(s)
Genetic Complementation Test , Hepacivirus/physiology , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/genetics , Virus Replication , Alleles , Cell Line , DNA Mutational Analysis , Hepacivirus/genetics , Hepatocytes/virology , Humans , Phosphorylation , Protein Processing, Post-Translational , Serine/genetics , Serine/metabolism
12.
Virology ; 422(1): 6-12, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22018491

ABSTRACT

Rift Valley fever virus (RVFV) causes significant morbidity and mortality in humans and livestock throughout Africa and the Middle East. The clinical disease ranges from mild febrile illness, to hepatitis, retinitis, encephalitis and fatal hemorrhagic fever. RVFV NSs protein has previously been shown to interfere in vitro with the interferon response, and RVFV lacking the NSs protein is attenuated in several animal models. Monocytes and macrophages are key players in the innate immune response via expression of various cytokines and chemokines. Here we demonstrate that wild-type RVFV infection of human monocyte-derived macrophages leads to a productive infection and inhibition of the innate immune response via decreased expression of IFN-α2, IFN-ß and TNF-α. Using a recombinant virus lacking the NSs protein, we show that this effect is mediated by the viral NSs protein. Finally, analysis of RVF patient samples demonstrated an association between a pro-inflammatory cytokine response and patient survival.


Subject(s)
Cytokines/blood , Macrophages/immunology , Macrophages/virology , Rift Valley Fever/immunology , Rift Valley fever virus , Viral Nonstructural Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cytokines/biosynthesis , Female , Humans , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Male , Middle Aged , RNA, Viral/genetics , RNA, Viral/metabolism , Rift Valley Fever/mortality , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Rift Valley fever virus/immunology , Rift Valley fever virus/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Viral Nonstructural Proteins/deficiency
13.
J Virol ; 85(19): 10058-68, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21752905

ABSTRACT

Previous studies have demonstrated that mouse hepatitis virus (MHV) hepatotropism is determined largely by postentry events rather than by availability of the viral receptor. In addition, mutation of MHV nonstructural protein 2 (ns2) abrogates the ability of the virus to replicate in the liver and induce hepatitis but does not affect replication in the central nervous system (CNS). Here we show that replication of ns2 mutant viruses is attenuated in bone marrow-derived macrophages (BMM) generated from wild-type (wt) mice but not in L2 fibroblasts, primary astrocytes, or BMM generated from type I interferon receptor-deficient (IFNAR(-/-)) mice. In addition, ns2 mutants are more sensitive than wt virus to pretreatment of BMM, but not L2 fibroblasts or primary astrocytes, with alpha/beta interferon (IFN-α/ß). The ns2 mutants induced similar levels of IFN-α/ß in wt and IFNAR(-/-) BMM, indicating that ns2 expression has no effect on the induction of IFN but rather that it antagonizes a later step in IFN signaling. Consistent with these in vitro data, the virulence of ns2 mutants increased to near that of wt virus after depletion of macrophages in vivo. These data imply that the ability of MHV to replicate in macrophages is a prerequisite for replication in the liver and induction of hepatitis but not for replication or disease in the CNS, underscoring the importance of IFN signaling in macrophages in vivo for protection of the host from hepatitis. Our results further support the notion that viral tissue tropism is determined in part by postentry events, including the early type I interferon response.


Subject(s)
Interferon Type I/immunology , Murine hepatitis virus/immunology , Murine hepatitis virus/physiology , Viral Tropism , Animals , Cells, Cultured , Coronavirus Infections/pathology , Coronavirus Infections/virology , Fibroblasts/virology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Leukocyte Reduction Procedures , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Rodent Diseases/pathology , Rodent Diseases/virology , Viral Nonstructural Proteins/deficiency
14.
Virol J ; 8: 240, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21595878

ABSTRACT

BACKGROUND: RAS, coded by ras proto-oncogenes, played an important role in signal transmission to regulate cell growth and differentiation. Host activation of RAS was significant for IFN-sensitive vaccinia virus (delE3L) or attenuate influenza virus in unallowable cells. RESULTS: Huamn NRAS gene was activated by mutating in codon 61. Then the activation of NRAS was detected by western blot in MDCK cells. The delNS1 H5N1 influenza virus with deletion of NS1 eIF4GI binding domain was weak multiplication in MDCK cells. And the replication of delNS1 virus and expression of IFN-beta and IRF-3 were detected by Real-time PCR in MDCK cells infected with delNS1 virus. It was found that the delNS1 virus had a significant increase in MDCK cells when the NRAS was activated, and yet, expression of IRF-3 and IFN-beta were restrained. CONCLUSIONS: The study demonstrated that activated NRAS played an important part for delNS1 virus replication in MDCK cells. Activated NRAS might be down-regulating the expression of antiviral cellular factors in delNS1 virus infected cells.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/growth & development , Viral Nonstructural Proteins/deficiency , ras Proteins/metabolism , Animals , Cell Line , Dogs , Humans , Influenza A Virus, H5N1 Subtype/genetics
15.
Virology ; 410(2): 375-84, 2011 Feb 20.
Article in English | MEDLINE | ID: mdl-21193212

ABSTRACT

MVM NS2 is essential for viral DNA amplification, but its mechanism of action is unknown. A classification scheme for autonomous parvovirus-associated replication (APAR) center development, based on NS1 distribution, was used to characterize abnormal APAR body maturation in NS2null mutant infections, and their organization examined for defects in host protein recruitment. Since acquisition of known replication factors appeared normal, we looked for differences in invoked DNA damage responses. We observed widespread association of H2AX/MDC1 damage response foci with viral replication centers, and sequestration and complex hyperphosphorylation of RPA(32), which occurred in wildtype and mutant infections. Quantifying these responses by western transfer indicated that both wildtype and NS2 mutant MVM elicited ATM activation, while phosphorylation of ATR, already basally activated in asynchronous A9 cells, was downregulated. We conclude that MVM infection invokes multiple damage responses that influence the APAR environment, but that NS2 does not modify the recruitment of cellular proteins.


Subject(s)
DNA Replication , Host-Pathogen Interactions , Minute Virus of Mice/physiology , Proteins/metabolism , Viral Nonstructural Proteins/deficiency , Virus Replication , Animals , Cell Line , DNA Repair , DNA, Viral/genetics , DNA, Viral/metabolism , Mice , Minute Virus of Mice/genetics
16.
Virus Res ; 154(1-2): 77-85, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20673840

ABSTRACT

Prior studies on PRRSV strain VR-2332 non-structural protein 2 (nsp2) had shown that as much as 403 amino acids could be removed from the hypervariable region without losing virus viability in vitro. We utilized selected nsp2 deletion mutants to examine in vivo growth. Young swine (4 pigs/group; 5 control swine) were inoculated intramuscularly with one of 4 nsp2 deletion mutants (rΔ727-813, rΔ543-726, rΔ324-523, rΔ324-726) or full-length recombinant virus (rVR-2332). Serum samples were collected on various days post-inoculation and analyzed by HerdChek* ELISA, PRRSV real time RT-PCR, gamma interferon (IFN-γ) ELISA, and nucleotide sequence analysis of the entire nsp2 coding region. Tracheobronchial lymph node weight compared to body weight was recorded for each animal and used as a clinical measurement of viral pathogenesis. Results showed that all deletion mutants grew less robustly than full-length recombinant virus, yet all but the large deletion virus (rΔ324-726) recovered to parental viral RNA levels by study end. Swine receiving the rΔ727-813 mutants had a significant decrease in lymph node enlargement compared to rVR-2332. While swine infection with rVR-2332 caused a rapid rise in serum IFN-γ levels, the IFN-γ protein produced by infection with 3 of the 4 deletion mutant viruses was significantly reduced, perhaps due to differences in viral growth kinetics. The rΔ543-726 nsp2 mutant virus, although growth impaired, mimicked rVR-2332 in inducing a host serum IFN-γ response but exhibited a 2-week delay. Targeted sequencing showed that all deletions were stable in the region coding for nsp2 after one swine passage. The data suggested that the selected nsp2 deletion mutants were growth attenuated in swine, altered the induction of serum IFN-γ, an innate cytokine of unknown function in PRRSV clearance, and pointed to a domain that may influence tracheobronchial lymph node size.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/pathology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/genetics , Sequence Deletion , Viral Nonstructural Proteins/deficiency , Animals , Body Weight , Interferon-gamma/blood , Lymph Nodes/pathology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Serum/virology , Swine , Time Factors , Virulence Factors/deficiency
17.
Bing Du Xue Bao ; 26(3): 244-8, 2010 May.
Article in Chinese | MEDLINE | ID: mdl-20572348

ABSTRACT

Based on the NSP4 sequence of bovine rotavirus (BRV), the shRNA was designed and synthesized, and a shRNA recombinant lenti-virus vector RNAi-H1-89 was constructed. The recombinant RNAi-H1-89 Lenti-virus was packaged by transfecting the 293T cell with the recombinant vector RNAi-H1-89 and two helper plasmids using lipofectamine, and then used to infect MA104 cells. The MA104 cells were further infected with BRV strain G6 24h post-infection, with the LacZ shRNA recombinant lenti-virus as control. Thirty-six hours later, the CPE of the infected cells was observed under microscope, shRNA of NSP4 gene inhibited CPE in MA104 cell; the shRNA against NSP4 gene also inhibited NSP4 gene expression by RT-PCR, The virus titer in the cell culture supernatant was significant lower compared with the control group. The above results showed that RNAi-H1-89 against NSP4 gene could specifically silence NSP4 gene expression, and inhibit the proliferation of BRV.


Subject(s)
Glycoproteins/deficiency , Glycoproteins/genetics , RNA, Small Interfering/genetics , Rotavirus/genetics , Rotavirus/physiology , Toxins, Biological/genetics , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/genetics , Virus Replication/genetics , Animals , Base Sequence , Cattle , Cell Line , DNA, Recombinant/genetics , Molecular Sequence Data , Plasmids/genetics , Viral Load/genetics
18.
Vaccine ; 27(34): 4622-9, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19523909

ABSTRACT

Integrase (IN) defective lentiviral vectors have a high safety profile and might prove useful as immunizing agents especially against HIV-1. However, IN defective SIV-based vectors must be developed in order to test their potential in the non-human primate models (NHP) of AIDS. To this aim we tested a novel SIV-based IN defective lentiviral vector for its ability to induce sustained immune responses in mice. BALB/c mice were immunized once intramuscularly with a SIV-based IN defective lentiviral vector expressing the model antigen enhanced green fluorescence protein (eGFP). Immune responses were evaluated 90 days after the injection and compared with those elicited with the IN competent counterpart. The IN defective vector was able to efficiently elicit specific and long-lasting polyfunctional immune responses as evaluated by enzyme-linked immunospot (ELISPOT) assays for interferon-gamma (IFN-gamma) in spleens, bone marrow (BM) and draining lymph nodes, and by intracellular staining (ICS) for IFN-gamma, Interleukin-2 (IL-2) and tumor necrosis factor (TNF-alpha) in both splenocytes and BM cells without integration of the vector into the host genome. This is the first demonstration that an IN defective SIV-based lentiviral vector provides effective immunization, thus paving the way for the construction of IN defective vectors expressing SIV antigen(s) and test their efficacy against a SIV virus challenge in the NHP model of AIDS.


Subject(s)
Genetic Vectors , Integrases/deficiency , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Viral Nonstructural Proteins/deficiency , Animals , Bone Marrow/immunology , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Leukocytes, Mononuclear/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Spleen/immunology , Tumor Necrosis Factor-alpha/immunology
19.
Wei Sheng Wu Xue Bao ; 48(12): 1666-70, 2008 Dec.
Article in Chinese | MEDLINE | ID: mdl-19271543

ABSTRACT

UNLABELLED: Infectious bursal disease virus (IBDV) belongs to genus Avibirnavirus of the family Birnaviridae. The genome of IBDV consists of two segments of double-strand RNA, which encode four structural protein VP1-VP4 and one non-structural protein VP5. OBJECTIVE: To study the function of VP5 of IBDV, the recombinant virus, lack of VP5 gene, was constructed and rescused by reverse genetic technique. METHODS: We deleted the VP5 gene (on segment A) of IBDV Gt strain by silence the start codon (ATG-ATC) using site-directed mutagenesis. The full length cDNA of segment A was flanked by hammerhead ribozyme and hepatitis delta virus ribozyme, which was introduced into an eukaryotic expression vector PCAGGS, under the strong chicken beta actin promoter. The recombinant plasmid was named as pCAGGmGtA deltaVP5HRT. Co-transfection was carried with PCAGGmGtAdVP5HRT and PCAGGmGtBHRT in DF-I cells. RESULTS: Recombinant virus was successfully rescused, which was verified by RT-PCR and indirect immnuofluorescence assay. The rescusd virus could be a very helpful platform for further study of VP5 biological function.


Subject(s)
Infectious bursal disease virus/genetics , Viral Nonstructural Proteins/deficiency , Virus Replication/genetics , Animals , Chickens , Chlorocebus aethiops , Infectious bursal disease virus/physiology , Mutagenesis, Site-Directed , Reverse Transcriptase Polymerase Chain Reaction , Viral Nonstructural Proteins/genetics
20.
J Gen Virol ; 88(Pt 5): 1403-1409, 2007 May.
Article in English | MEDLINE | ID: mdl-17412966

ABSTRACT

Non-structural protein NS1 of influenza A virus counteracts the host immune response by blocking the synthesis of type I interferon (IFN). As deletion of the complete NS1 gene has to date been reported only in the human H1N1 strain A/PR/8/34, it remained unclear whether NS1 is a non-essential virulence factor in other influenza A virus strains as well. In this report, the properties of NS1-deficient mutants derived from strain SC35M (H7N7) are described. A mutant of SC35M that completely lacks the NS1 gene was an excellent inducer of IFN in mammalian and avian cells in culture and, consequently, was able to multiply efficiently only in cell lines with defects in the type I IFN system. Virus mutants carrying C-terminally truncated versions of NS1 were less powerful inducers of IFN and were attenuated less strongly in human A549 cells. Although attenuated in wild-type mice, these mutants remained highly pathogenic for mice lacking the IFN-regulated antiviral factor Mx1. In contrast, the NS1-deficient SC35M mutant was completely non-pathogenic for wild-type mice, but remained pathogenic for mice lacking Mx1 and double-stranded RNA-activated protein kinase (PKR). Wild-type SC35M, but not the NS1-deficient mutant virus, was able to replicate in the upper respiratory tract of birds, but neither virus induced severe disease in adult chickens. Altogether, this study supports the view that NS1 represents a non-essential virulence factor of different influenza A viruses.


Subject(s)
Influenza A Virus, H7N7 Subtype/physiology , Influenza in Birds/immunology , Viral Nonstructural Proteins/deficiency , Animals , Cell Line , Chickens , Chlorocebus aethiops , Dogs , Humans , Influenza A Virus, H7N7 Subtype/classification , Influenza A Virus, H7N7 Subtype/isolation & purification , Influenza, Human , Interferon-beta/genetics , Interferons/antagonists & inhibitors , Lung/immunology , Lung/virology , Mice , Orthomyxoviridae Infections , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...