Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
mBio ; 13(5): e0186122, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36102514

ABSTRACT

Understanding the complex interactions between virus and host that drive new strain evolution is key to predicting the emergence potential of variants and informing vaccine development. Under our hypothesis, future dominant human norovirus GII.4 variants with critical antigenic properties that allow them to spread are currently circulating undetected, having diverged years earlier. Through large-scale sequencing of GII.4 surveillance samples, we identified two variants with extensive divergence within domains that mediate neutralizing antibody binding. Subsequent serological characterization of these strains using temporally resolved adult and child sera suggests that neither candidate could spread globally in adults with multiple GII.4 exposures, yet young children with minimal GII.4 exposure appear susceptible. Antigenic cartography of surveillance and outbreak sera indicates that continued population exposure to GII.4 Sydney 2012 and antigenically related variants over a 6-year period resulted in a broadening of immunity to heterogeneous GII.4 variants, including those identified here. We show that the strongest antibody responses in adults exposed to GII.4 Sydney 2012 are directed to previously circulating GII.4 viruses. Our data suggest that the broadening of antibody responses compromises establishment of strong GII.4 Sydney 2012 immunity, thereby allowing the continued persistence of GII.4 Sydney 2012 and modulating the cycle of norovirus GII.4 variant replacement. Our results indicate a cycle of norovirus GII.4 variant replacement dependent upon population immunity. Young children are susceptible to divergent variants; therefore, emergence of these strains worldwide is driven proximally by changes in adult serological immunity and distally by viral evolution that confers fitness in the context of immunity. IMPORTANCE In our model, preepidemic human norovirus variants harbor genetic diversification that translates into novel antigenic features without compromising viral fitness. Through surveillance, we identified two viruses fitting this profile, forming long branches on a phylogenetic tree. Neither evades current adult immunity, yet young children are likely susceptible. By comparing serological responses, we demonstrate that population immunity varies by age/exposure, impacting predicted susceptibility to variants. Repeat exposure to antigenically similar variants broadens antibody responses, providing immunological coverage of diverse variants but compromising response to the infecting variant, allowing continued circulation. These data indicate norovirus GII.4 variant replacement is driven distally by virus evolution and proximally by immunity in adults.


Subject(s)
Caliciviridae Infections , Norovirus , Adult , Child , Humans , Child, Preschool , Phylogeny , Antibodies, Neutralizing , Disease Outbreaks/prevention & control , Genotype
3.
Euro Surveill ; 25(43)2020 10.
Article in English | MEDLINE | ID: mdl-33124554

ABSTRACT

BackgroundRapid diagnostic tests are commonly used by hospital laboratories in England to detect rotavirus (RV), and results are used to inform clinical management and support national surveillance of the infant rotavirus immunisation programme since 2013. In 2017, the Public Health England (PHE) national reference laboratory for enteric viruses observed that the presence of RV could not be confirmed by PCR in a proportion of RV-positive samples referred for confirmatory detection.AimWe aimed to compare the positivity rate of detection methods used by hospital laboratories with the PHE confirmatory test rate.MethodsRotavirus specimens testing positive at local hospital laboratories were re-tested at the PHE national reference laboratory using a PCR test. Confirmatory results were compared to original results from the PHE laboratory information management system.ResultsHospital laboratories screened 70.1% (2,608/3,721) of RV samples using immunochromatographic assay (IC) or rapid tests, 15.5% (578/3,721) using enzyme immunoassays (EIA) and 14.4% (535/3,721) using PCR. Overall, 1,011/3,721 (27.2%) locally RV-positive samples referred to PHE in 2016 and 2017 failed RV detection using the PHE reference laboratory PCR test. Confirmation rates were 66.9% (1,746/2,608) for the IC tests, 87.4% (505/578) for the EIA and 86.4% (465/535) for the PCR assays. Seasonal confirmation rate discrepancies were also evident for IC tests.ConclusionsThis report highlights high false positive rates with the most commonly used RV screening tests and emphasises the importance of implementing verified confirmatory tests for RV detections. This has implications for clinical diagnosis and national surveillance.


Subject(s)
Public Health Surveillance , Rotavirus Infections , Rotavirus , England/epidemiology , Humans , Infant , Retrospective Studies , Rotavirus/isolation & purification , Rotavirus Infections/diagnosis , Rotavirus Infections/epidemiology
4.
Nat Commun ; 10(1): 2216, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101900

ABSTRACT

Transcribing and replicating a double-stranded genome require protein modules to unwind, transcribe/replicate nucleic acid substrates, and release products. Here we present in situ cryo-electron microscopy structures of rotavirus dsRNA-dependent RNA polymerase (RdRp) in two states pertaining to transcription. In addition to the previously discovered universal "hand-shaped" polymerase core domain shared by DNA polymerases and telomerases, our results show the function of N- and C-terminal domains of RdRp: the former opens the genome duplex to isolate the template strand; the latter splits the emerging template-transcript hybrid, guides genome reannealing to form a transcription bubble, and opens a capsid shell protein (CSP) to release the transcript. These two "helicase" domains also extensively interact with CSP, which has a switchable N-terminal helix that, like cellular transcriptional factors, either inhibits or promotes RdRp activity. The in situ structures of RdRp, CSP, and RNA in action inform mechanisms of not only transcription, but also replication.


Subject(s)
DNA Replication/physiology , RNA, Messenger/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Rotavirus/physiology , Transcription, Genetic/physiology , Animals , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Cell Line , Chlorocebus aethiops , Cryoelectron Microscopy , Models, Molecular , Protein Domains/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Rotavirus/ultrastructure , Virus Replication/physiology
5.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30518645

ABSTRACT

Bluetongue virus (BTV), in the family Reoviridae, is an insect-borne, double-capsid virus causing hemorrhagic disease in livestock around the world. Here, we elucidate how outer capsid proteins VP2 and VP5 coordinate cell entry of BTV. To identify key functional residues, we used atomic-level structural data to guide mutagenesis of VP2 and VP5 and a series of biological and biochemical approaches, including site-directed mutagenesis, reverse genetics-based virus recovery, expression and characterization of individual recombinant mutant proteins, and various in vitro and in vivo assays. We demonstrate the dynamic nature of the conformational change process, revealing that a unique zinc finger (CCCH) in VP2 acts as the major low pH sensor, coordinating VP2 detachment, subsequently allowing VP5 to sense low pH via specific histidine residues at key positions. We show that single substitution of only certain histidine residues has a lethal effect, indicating that the location of histidine in VP5 is critical to inducing changes in VP5 conformation that facilitates membrane penetration. Further, we show that the VP5 anchoring domain alone recapitulates sensing of low pH. Our data reveal a novel, multiconformational process that overcomes entry barriers faced by this multicapsid nonenveloped virus.IMPORTANCE Virus entry into a susceptible cell is the first step of infection and a significant point at which infection can be prevented. To enter effectively, viruses must sense the cellular environment and, when appropriate, initiate a series of changes that eventually jettison the protective shell and deposit virus genes into the cytoplasm. Many viruses sense pH, but how this happens and the events that follow are often poorly understood. Here, we address this question for a large multilayered bluetongue virus. We show key residues in outer capsid proteins, a pH-sensing histidine of a zinc finger within the receptor-binding VP2 protein, and certain histidine residues in the membrane-penetrating VP5 protein that detect cellular pH, leading to irreversible changes and propel the virus through the cell membrane. Our data reveal a novel mechanism of cell entry for a nonenveloped virus and highlight mechanisms which may also be used by other viruses.


Subject(s)
Bluetongue virus/genetics , Bluetongue virus/metabolism , Host Microbial Interactions/physiology , Bluetongue virus/pathogenicity , Capsid Proteins/genetics , Cell Line , Cell Membrane/metabolism , DNA Viruses/genetics , Hydrogen-Ion Concentration , Protein Binding/physiology , Reoviridae/genetics , Virion/genetics , Virus Internalization
6.
Viruses ; 9(7)2017 06 29.
Article in English | MEDLINE | ID: mdl-28661470

ABSTRACT

Rotavirus (RV), a member of the Reoviridae family, causes infection in children and infants, with high morbidity and mortality. To be viable, the virus particle must package a set of eleven RNA segments. In order to understand the packaging mechanism, here, we co-synthesized sets of RNA segments in vitro in different combinations and detected by two alternate methods: the electrophoretic mobility shift assay (EMSA) and the RNA-bead pull-down assay. We showed that viral positive-sense RNA segments interact with each other in a specific manner, forming RNA complexes, and that the RNA-RNA interactions followed a sequential order initiated by small RV segments. Further, we demonstrated that RNA complexes were perturbed by targeted specific antisense oligoribonucleotides (ORNs) complementary to short RNA sequences, indicating that the RNA-RNA interactions between different segments were sequence-specific. The same inhibitory ORNs also had the capability to inhibit virus replication. The combined in vitro and in vivo data inferred that RNA-RNA interactions and specific complex formation are essential for sorting different segments, possibly prior to, or during, genome packaging. As genome assembly is a universal requirement in the Reoviridae family, this work offers an approach towards a further understanding of the sorting and packaging mechanisms of RV and related dsRNA (double-stranded RNA) viruses.


Subject(s)
RNA, Viral/metabolism , Rotavirus/physiology , Virus Assembly , Animals , Cell Line , Chlorocebus aethiops , Macromolecular Substances/metabolism , RNA, Viral/genetics , Rotavirus/genetics
7.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795442

ABSTRACT

Bluetongue virus (BTV) is endemic in many parts of the world, often causing severe hemorrhagic disease in livestock. To date, at least 27 different serotypes have been recognized. Vaccination against all serotypes is necessary to protect susceptible animals and to prevent onward spread of the virus by insect vectors. In our previous studies, we generated replication-deficient (disabled infectious single-cycle [DISC]) virus strains for a number of serotypes and reported preliminary data on their protective efficacy in animals. In this report, to advance the DISC vaccines to the marketplace, we investigated different parameters of these DISC vaccines. First, we demonstrated the genetic stabilities of these vaccine strains and also the complementing cell line. Subsequently, the optimal storage conditions of vaccines, including additives, temperature, and desiccation, were determined and their protective efficacies in animals confirmed. Furthermore, to test if mixtures of different vaccine strains could be tolerated, we tested cocktails of DISC vaccines in combinations of three or six different serotypes in sheep and cattle, the two natural hosts of BTV. Groups of sheep vaccinated with a cocktail of six different vaccines were completely protected from challenge with individual virulent serotypes, both in early challenge and after 5 months of challenge without any clinical disease. There was no interference in protection between the different vaccines. Protection was also achieved in cattle with a mixture of three vaccine strains, albeit at a lesser level than in sheep. Our data support and validate the suitability of these virus strains as the next-generation vaccines for BTV. IMPORTANCE: Bluetongue (BT) is a debilitating and in many cases lethal disease that affects ruminants of economic importance. Classical vaccines that afford protection against bluetongue virus, the etiological agent, are not free from secondary and undesirable effects. A surge in new approaches to produce highly attenuated, safer vaccines was evident after the development of the BTV reverse-genetics system that allows the introduction of targeted mutations in the virus genome. We targeted an essential gene to develop disabled virus strains as vaccine candidates. The results presented in this report further substantiate our previous evidence and support the suitability of these virus strains as the next-generation BTV vaccines.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Bluetongue virus/drug effects , Bluetongue/prevention & control , Viral Vaccines/immunology , Virion/immunology , Animals , Base Sequence , Bluetongue/immunology , Bluetongue/virology , Bluetongue virus/classification , Bluetongue virus/genetics , Bluetongue virus/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Cattle , Cell Line , Drug Stability , Drug Storage , Female , Male , Reverse Genetics , Serogroup , Sheep , Vaccination , Vaccines, Attenuated , Vaccines, Subunit , Viral Vaccines/administration & dosage , Viral Vaccines/biosynthesis , Viral Vaccines/genetics , Virion/genetics
8.
Nat Struct Mol Biol ; 23(1): 74-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26641711

ABSTRACT

Viruses sense environmental cues such as pH to engage in membrane interactions for cell entry during infection, but how nonenveloped viruses sense pH is largely undefined. Here, we report both high- and low-pH structures of bluetongue virus (BTV), which enters cells via a two-stage endosomal process. The receptor-binding protein VP2 possesses a zinc finger that may function to maintain VP2 in a metastable state and a conserved His866, which senses early-endosomal pH. The membrane-penetration protein VP5 has three domains: dagger, unfurling and anchoring. Notably, the ß-meander motif of the anchoring domain contains a histidine cluster that can sense late-endosomal pH and also possesses four putative membrane-interaction elements. Exposing BTV to low pH detaches VP2 and dramatically refolds the dagger and unfurling domains of VP5. Our biochemical and structure-guided-mutagenesis studies support these coordinated pH-sensing mechanisms.


Subject(s)
Bluetongue virus/drug effects , Bluetongue virus/physiology , Virus Internalization/drug effects , Amino Acid Motifs , Bluetongue virus/chemistry , Capsid Proteins/chemistry , Capsid Proteins/metabolism , DNA Mutational Analysis , Endocytosis , Hydrogen-Ion Concentration , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation/drug effects , Zinc Fingers
9.
Viruses ; 7(5): 2378-403, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25984713

ABSTRACT

Bluetongue virus (BTV), a non-enveloped arbovirus, causes hemorrhagic disease in ruminants. However, the influence of natural host cell proteins on BTV replication process is not defined. In addition to cell lysis, BTV also exits non-ovine cultured cells by non-lytic pathways mediated by nonstructural protein NS3 that interacts with virus capsid and cellular proteins belonging to calpactin and ESCRT family. The PPXY late domain motif known to recruit NEDD4 family of HECT ubiquitin E3 ligases is also highly conserved in NS3. In this study using a mixture of molecular, biochemical and microscopic techniques we have analyzed the importance of ovine cellular proteins and vesicles in BTV infection. Electron microscopic analysis of BTV infected ovine cells demonstrated close association of mature particles with intracellular vesicles. Inhibition of Multi Vesicular Body (MVB) resident lipid phosphatidylinositol-3-phosphate resulted in decreased total virus titre suggesting that the vesicles might be MVBs. Proteasome mediated inhibition of ubiquitin or modification of virus lacking the PPXY in NS3 reduced virus growth. Thus, our study demonstrated that cellular components comprising of MVB and exocytic pathways proteins are involved in BTV replication in ovine cells.


Subject(s)
Bluetongue virus/physiology , Host-Pathogen Interactions , Virus Replication , Animals , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/virology , Protein Transport , Sheep , Viral Proteins/metabolism
10.
Vet Microbiol ; 174(1-2): 139-47, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25307940

ABSTRACT

Bluetongue (BT) disease, caused by the non-enveloped bluetongue virus (BTV) belonging to the Reoviridae family, is an economically important disease that affects a wide range of wild and domestic ruminants. Currently, 26 different serotypes of BTV are recognized in the world, of which BTV-8 has been found to exhibit one of the most virulent manifestations of BT disease in livestock. In recent years incursions of BTV-8 in Europe have resulted in significant morbidity and mortality not only in sheep but also in cattle. The molecular and genetic basis of BTV-8 pathogenesis is not known. To understand the genetic basis of BTV-8 pathogenicity, we generated reassortant viruses by replacing the 3 most variable genes, S2, S6 and S10 of a recent isolate of BTV-8, in different combinations into the backbone of an attenuated strain of BTV-1. The growth profiles of these reassortant viruses were then analyzed in two different ovine cell lines derived from different organs, kidney and thymus. Distinct patterns for each reassortant virus in these two cell lines were observed. To determine the pathogenicity of these reassortant viruses, groups of BTV-susceptible sheep were infected with each of these viruses. The data suggested that the clinical manifestations of these two different serotypes, BTV-1 and BTV-8, were slightly distinct and BTV-1, when comprising all 3 genome segments of BTV-8, behaved differently to BTV-1. Our results also suggested that the molecular basis of BT disease is highly complex.


Subject(s)
Bluetongue virus/pathogenicity , Bluetongue/virology , Reassortant Viruses/pathogenicity , Reverse Genetics/veterinary , Animals , Bluetongue virus/genetics , Bluetongue virus/growth & development , Cell Line , Enzyme-Linked Immunosorbent Assay/veterinary , Reassortant Viruses/genetics , Reverse Genetics/methods , Serogroup , Sheep , Viral Load/veterinary , Virulence/genetics
11.
J Gen Virol ; 95(Pt 10): 2240-2250, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24980574

ABSTRACT

The mechanism used by bluetongue virus (BTV) to ensure the sorting and packaging of its 10 genomic segments is still poorly understood. In this study, we investigated the packaging constraints for two BTV genomic segments from two different serotypes. Segment 4 (S4) of BTV serotype 9 was mutated sequentially and packaging of mutant ssRNAs was investigated by two newly developed RNA packaging assay systems, one in vivo and the other in vitro. Modelling of the mutated ssRNA followed by biochemical data analysis suggested that a conformational motif formed by interaction of the 5' and 3' ends of the molecule was necessary and sufficient for packaging. A similar structural signal was also identified in S8 of BTV serotype 1. Furthermore, the same conformational analysis of secondary structures for positive-sense ssRNAs was used to generate a chimeric segment that maintained the putative packaging motif but contained unrelated internal sequences. This chimeric segment was packaged successfully, confirming that the motif identified directs the correct packaging of the segment.


Subject(s)
Bluetongue virus/physiology , RNA, Viral/metabolism , Virus Assembly , Bluetongue virus/genetics , DNA Mutational Analysis , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Viral/genetics , Sequence Analysis, DNA
12.
J Virol ; 87(17): 9856-64, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824810

ABSTRACT

Since 1998, 9 of the 26 serotypes of bluetongue virus (BTV) have spread throughout Europe, and serotype 8 has suddenly emerged in northern Europe, causing considerable economic losses, direct (mortality and morbidity) but also indirect, due to restriction in animal movements. Therefore, many new types of vaccines, particularly subunit vaccines, with improved safety and efficacy for a broad range of BTV serotypes are currently being developed by different laboratories. Here we exploited a reverse genetics-based replication-deficient BTV serotype 1 (BTV-1) (disabled infectious single cycle [DISC]) strain to generate a series of DISC vaccine strains. Cattle and sheep were vaccinated with these viruses either singly or in cocktail form as a multivalent vaccine candidate. All vaccinated animals were seroconverted and developed neutralizing antibody responses to their respective serotypes. After challenge with the virulent strains at 21 days postvaccination, vaccinated animals showed neither any clinical reaction nor viremia. Further, there was no interference with protection with a multivalent preparation of six distinct DISC viruses. These data indicate that a very-rapid-response vaccine could be developed based on which serotypes are circulating in the population at the time of an outbreak.


Subject(s)
Bluetongue virus/immunology , Bluetongue/prevention & control , Cattle Diseases/prevention & control , Viral Vaccines/isolation & purification , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Bluetongue/epidemiology , Bluetongue/immunology , Bluetongue virus/classification , Bluetongue virus/genetics , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/immunology , Disease Outbreaks/prevention & control , Disease Outbreaks/veterinary , Europe/epidemiology , Female , RNA, Viral/genetics , Reassortant Viruses/genetics , Reassortant Viruses/immunology , Serotyping , Sheep, Domestic , Vaccines, Subunit/isolation & purification , Virus Replication/genetics
13.
J Gen Virol ; 94(Pt 10): 2259-2265, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23860489

ABSTRACT

African horse sickness virus (AHSV) is an insect-vectored emerging pathogen of equine species. AHSV (nine serotypes) is a member of the genus Orbivirus, with a morphology and coding strategy similar to that of the type member, bluetongue virus. However, these viruses are distinct at the genetic level, in the proteins they encode and in their pathobiology. AHSV infection of horses is highly virulent with a mortality rate of up to 90 %. AHSV is transmitted by Culicoides, a common European insect, and has the potential to emerge in Europe from endemic countries of Africa. As a result, a safe and effective vaccine is sought urgently. As part of a programme to generate a designed highly attenuated vaccine, we report here the recovery of AHSV from a complete set of RNA transcripts synthesized in vitro from cDNA clones. We have demonstrated the generation of mutant and reassortant AHSV genomes, their recovery, stable passage, and characterization. Our findings provide a new approach to investigate AHSV replication, to design AHSV vaccines and to aid diagnosis.


Subject(s)
African Horse Sickness Virus/genetics , African Horse Sickness Virus/metabolism , RNA, Viral/biosynthesis , RNA, Viral/chemistry , Animals , Cell Line , Cloning, Molecular , Cricetinae , Gene Expression Regulation, Viral/physiology , Genome, Viral , RNA, Viral/genetics , Reassortant Viruses , Virus Replication
14.
Virol J ; 9: 178, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22931514

ABSTRACT

BACKGROUND: Bluetongue virus (BTV) is a double-stranded RNA (dsRNA) virus of the Reoviridae family, which encodes its genes in ten linear dsRNA segments. BTV mRNAs are synthesised by the viral RNA-dependent RNA polymerase (RdRp) as exact plus sense copies of the genome segments. Infection of mammalian cells with BTV rapidly replaces cellular protein synthesis with viral protein synthesis, but the regulation of viral gene expression in the Orbivirus genus has not been investigated. RESULTS: Using an mRNA reporter system based on genome segment 10 of BTV fused with GFP we identify the protein characteristic of this genus, non-structural protein 1 (NS1) as sufficient to upregulate translation. The wider applicability of this phenomenon among the viral genes is demonstrated using the untranslated regions (UTRs) of BTV genome segments flanking the quantifiable Renilla luciferase ORF in chimeric mRNAs. The UTRs of viral mRNAs are shown to be determinants of the amount of protein synthesised, with the pre-expression of NS1 increasing the quantity in each case. The increased expression induced by pre-expression of NS1 is confirmed in virus infected cells by generating a replicating virus which expresses the reporter fused with genome segment 10, using reverse genetics. Moreover, NS1-mediated upregulation of expression is restricted to mRNAs which lack the cellular 3' poly(A) sequence identifying the 3' end as a necessary determinant in specifically increasing the translation of viral mRNA in the presence of cellular mRNA. CONCLUSIONS: NS1 is identified as a positive regulator of viral protein synthesis. We propose a model of translational regulation where NS1 upregulates the synthesis of viral proteins, including itself, and creates a positive feedback loop of NS1 expression, which rapidly increases the expression of all the viral proteins. The efficient translation of viral reporter mRNAs among cellular mRNAs can account for the observed replacement of cellular protein synthesis with viral protein synthesis during infection.


Subject(s)
Bluetongue virus/metabolism , Gene Expression Regulation, Viral/physiology , Viral Nonstructural Proteins/metabolism , Animals , Bluetongue virus/genetics , Cell Line , Cricetinae , Genes, Reporter , Genome, Viral , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation , Viral Nonstructural Proteins/genetics
15.
J Virol ; 85(19): 10213-21, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795358

ABSTRACT

The reverse genetics technology for bluetongue virus (BTV) has been used in combination with complementing cell lines to recover defective BTV-1 mutants. To generate a potential disabled infectious single cycle (DISC) vaccine strain, we used a reverse genetics system to rescue defective virus strains with large deletions in an essential BTV gene that encodes the VP6 protein (segment S9) of the internal core. Four VP6-deficient BTV-1 mutants were generated by using a complementing cell line that provided the VP6 protein in trans. Characterization of the growth properties of mutant viruses showed that each mutant has the necessary characteristics for a potential vaccine strain: (i) viral protein expression in noncomplementing mammalian cells, (ii) no infectious virus generated in noncomplementing cells, and (iii) efficient replication in the complementing VP6 cell line. Further, a defective BTV-8 strain was made by reassorting the two RNA segments that encode the two outer capsid proteins (VP2 and VP5) of a highly pathogenic BTV-8 with the remaining eight RNA segments of one of the BTV-1 DISC viruses. The protective capabilities of BTV-1 and BTV-8 DISC viruses were assessed in sheep by challenge with specific virulent strains using several assay systems. The data obtained from these studies demonstrated that the DISC viruses are highly protective and could offer a promising alternative to the currently available attenuated and killed virus vaccines and are also compliant as DIVA (differentiating infected from vaccinated animals) vaccines.


Subject(s)
Bluetongue virus/immunology , Bluetongue/prevention & control , Defective Viruses/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Bluetongue virus/genetics , Bluetongue virus/isolation & purification , Cell Culture Techniques , Defective Viruses/genetics , Defective Viruses/isolation & purification , Female , Male , Reassortant Viruses/genetics , Reassortant Viruses/immunology , Reassortant Viruses/isolation & purification , Sheep , Viral Vaccines/genetics , Viral Vaccines/isolation & purification , Viremia/prevention & control
16.
J Virol ; 85(10): 4783-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21411520

ABSTRACT

Bluetongue virus (BTV), a member of the Reoviridae family, is an insect-borne animal pathogen. Virus release from infected cells is predominantly by cell lysis, but some BTV particles are also released from the plasma membrane. The nonstructural protein NS3 has been implicated in this process. Using alternate initiator methionine residues, NS3 is expressed as a full-length protein and as a truncated variant that lacks the initial 13 residues, which, by yeast-two hybrid analyses, have been shown to interact with a cellular trafficking protein S100A10/p11. To understand the physiological significance of this interaction in virus-infected cells, we have used reverse genetics to investigate the roles of NS3 and NS3A in virus replication and localization in both mammalian and insect vector-derived cells. A virus expressing NS3 but not NS3A was able to propagate in and release from mammalian cells efficiently. However, growth of a mutant virus expressing only NS3A was severely attenuated, although protein expression, replication, double-stranded RNA (dsRNA) synthesis, and particle assembly in the cytoplasm were observed. Two of three single-amino-acid substitutions in the N-terminal 13 residues of NS3 showed phenotypically similar effects. Pulldown assay and confocal microscopy demonstrated a lack of interaction between NS3 and S100A10/p11 in mutants with poor replication. The role of NS3/NS3A was also assessed in insect cells where virus grew, albeit with a reduced titer. Notably, however, while wild-type particles were found within cytoplasmic vesicles in insect cells, mutant viruses were scattered throughout the cytoplasm and not confined to vesicles. These results provide support for a role for the extreme amino terminus of NS3 in the late stages of virus growth in mammalian cells, plausibly in egress. However, both NS3 and NS3A were required for efficient BTV growth in insect cells.


Subject(s)
Annexin A2/metabolism , Bluetongue virus/pathogenicity , Host-Pathogen Interactions , Protein Interaction Mapping , S100 Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Amino Acid Substitution , Animals , Bluetongue virus/growth & development , Cell Line , Cricetinae , Gene Deletion , Insecta , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Two-Hybrid System Techniques , Viral Nonstructural Proteins/genetics
17.
FEBS Lett ; 584(15): 3386-91, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20600010

ABSTRACT

African horse sickness virus (AHSV), a member of the orbivirus genus of the family Reoviridae, is an insect-vectored pathogen of horses of concern to the equine industry. Studies on AHSV replication and pathogenesis have been hampered by the lack of reverse genetics allowing targeted mutation of viral genomes. We demonstrate that AHSV single-stranded RNA synthesized in vitro (core transcripts) is infectious and that there are distinct primary and secondary stages of the replication cycle. Transfection with a mixture of core transcripts from two different serotypes or a mixture of core transcripts and a T7 derived transcript resulted in the recovery of reassortant viruses. Recovery of infectious ASHV from nucleic acid will benefit investigation of the virus and the generation of attenuated vaccines.


Subject(s)
African Horse Sickness Virus/genetics , African Horse Sickness Virus/physiology , Genetic Techniques , Virus Replication/physiology , African Horse Sickness Virus/isolation & purification , African Horse Sickness Virus/ultrastructure , Animals , Cell Line , DNA, Complementary/genetics , Gene Expression Regulation, Viral , Genome, Viral/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , Reassortant Viruses/classification , Reassortant Viruses/genetics , Reassortant Viruses/physiology , Serotyping , Transfection , Virion/isolation & purification , Virion/ultrastructure
18.
BMC Mol Biol ; 10: 87, 2009 Sep 02.
Article in English | MEDLINE | ID: mdl-19725957

ABSTRACT

BACKGROUND: Many cellular multi-protein complexes are naturally present in cells at low abundance. Baculovirus expression offers one approach to produce milligram quantities of correctly folded and processed eukaryotic protein complexes. However, current strategies suffer from the need to produce large transfer vectors, and the use of repeated promoter sequences in baculovirus, which itself produces proteins that promote homologous recombination. One possible solution to these problems is to construct baculovirus genomes that express each protein in a complex from a separate locus within the viral DNA. However current methods for selecting such recombinant genomes are too inefficient to routinely modify the virus in this way. RESULTS: This paper reports a method which combines the lambda red and bacteriophage P1 Cre-recombinase systems to efficiently generate baculoviruses in which protein complexes are expressed from multiple, single-locus insertions of foreign genes. This method is based on an 88 fold improvement in the selection of recombinant viruses generated by red recombination techniques through use of a bipartite selection cassette. Using this system, seven new genetic loci were identified in the AcMNPV genome suitable for the high level expression of recombinant proteins. These loci were used to allow the recovery two recombinant virus-like particles with potential biotechnological applications (influenza A virus HA/M1 particles and bluetongue virus VP2/VP3/VP5/VP7 particles) and the mammalian chaperone and cancer drug target CCT (16 subunits formed from 8 proteins). CONCLUSION: 1. Use of bipartite selections can significantly improve selection of modified bacterial artificial chromosomes carrying baculovirus DNA. Furthermore this approach is sufficiently robust to allow routine modification of the virus genome. 2. In addition to the commonly used p10 and polyhedrin loci, the ctx, egt, 39k, orf51, gp37, iap2 and odv-e56 loci in AcMNPV are all suitable for the high level expression of heterologous genes. 3. Two protein, four protein and eight protein complexes including virus-like particles and cellular chaperone complexes can be produced using the new approach.


Subject(s)
Baculoviridae/genetics , Gene Expression , Genetic Engineering/methods , Genetic Vectors/genetics , Genome, Viral , Animals , Baculoviridae/metabolism , Chaperonin Containing TCP-1 , Chaperonins/genetics , Chaperonins/metabolism , Genetic Vectors/metabolism , Mice , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera
19.
J Virol ; 83(13): 6806-16, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19369335

ABSTRACT

Bluetongue virus (BTV), a nonenveloped insect-borne virus, is released from infected cells by multiple pathways. Unlike other nonenveloped viruses, in addition to cell lysis the newly synthesized virus particles also appear to use a unique "budding" process. The nonstructural protein NS3, the only membrane protein encoded by BTV in infected cells, has been implicated in this process, since it appears to interact not only with the outermost viral capsid protein VP2 but also with a component of the cellular ESCRT pathway. However, to date it had not been possible to obtain direct evidence for the involvement of NS3 in BTV morphogenesis due to the lack of a genetic system that would allow introducing the targeted mutation in NS3 gene. In this study, we have used the recently developed T7 transcript-based reverse genetics system for BTV to introduce mutations in the sequence of NS3 into the viral genome and have investigated the effect of these mutations in the context of a replicating virus. While certain NS3 mutations exhibited drastic effects on newly synthesized virus release, others had less pronounced effects. In particular, mutations of two residues in the Tsg101 binding motif, the putative L domain of NS3, altered normal virus egress patterns and left nascent particles tethered to the cellular membrane, apparently arrested in the process of budding. In cells infected with a mutant virus that was incapable of an NS3-VP2 interaction, no budding particles were visualized. These data suggest that NS3 may act like the membrane protein of enveloped viruses and is responsible for intracellular trafficking and budding of virus particles. NS3 is thus a bridge between the maturing virion particles and cellular proteins during virus egress.


Subject(s)
Bluetongue virus/physiology , Capsid Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Amino Acid Sequence , Animals , Bluetongue virus/genetics , Bluetongue virus/metabolism , Cell Line , Cricetinae , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Interaction Domains and Motifs , RNA, Viral/genetics , Viral Nonstructural Proteins/genetics
20.
Virol J ; 5: 82, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18638365

ABSTRACT

BACKGROUND: Studies on Rift Valley Fever Virus (RVFV) infection process and morphogenesis have been hampered due to the biosafety conditions required to handle this virus, making alternative systems such as recombinant virus-like particles, that may facilitate understanding of these processes are highly desirable. In this report we present the expression and characterization of RVFV structural proteins N, Gn and Gc and demonstrate the efficient generation of RVFV virus-like particles (VLPs) using a baculovirus expression system. RESULTS: A recombinant baculovirus, expressing nucleocapsid (N) protein of RVFV at high level under the control of the polyhedrin promoter was generated. Gel filtration analysis indicated that expressed N protein could form complex multimers. Further, N protein complex when visualized by electron microscopy (EM) exhibited particulate, nucleocapsid like-particles (NLPs). Subsequently, a single recombinant virus was generated that expressed the RVFV glycoproteins (Gn/Gc) together with the N protein using a dual baculovirus vector. Both the Gn and Gc glycoproteins were detected not only in the cytoplasm but also on the cell surface of infected cells. Moreover, expression of the Gn/Gc in insect cells was able to induce cell-cell fusion after a low pH shift indicating the retention of their functional characteristics. In addition, assembly of these three structural proteins into VLPs was identified by purification of cells' supernatant through potassium tartrate-glycerol gradient centrifugation followed by EM analysis. The purified particles exhibited enveloped structures that were similar to the structures of the wild-type RVFV virion particle. In parallel, a second recombinant virus was constructed that expressed only Gc protein together with N protein. This dual recombinant virus also generated VLPs with clear spiky structures, but appeared to be more pleomorphic than the VLPs with both glycoproteins, suggesting that Gc and probably also Gn interacts with N protein complex independent of each other. CONCLUSION: Our results suggest that baculovirus expression system has enormous potential to produce large amount of VLPs that may be used both for fundamental and applied research of RVFV.


Subject(s)
Recombinant Proteins/metabolism , Rift Valley fever virus/genetics , Viral Structural Proteins/metabolism , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Cells, Cultured , Glycoproteins/genetics , Glycoproteins/metabolism , Microscopy, Electron , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Recombinant Proteins/genetics , Rift Valley fever virus/metabolism , Spodoptera , Viral Structural Proteins/genetics , Virion/metabolism , Virology/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...