Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Nucleic Acids Res ; 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769067

RESUMO

How do segmented RNA viruses correctly recruit their genome has yet to be clarified. Bluetongue virus is a double-stranded RNA virus with 10 segments of different sizes, but it assembles its genome in single-stranded form through a series of specific RNA-RNA interactions prior to packaging. In this study, we determined the structure of each BTV transcript, individually and in different combinations, using 2'-hydroxyl acylation analysed by primer extension and mutational profiling (SHAPE-MaP). SHAPE-MaP identified RNA structural changes during complex formation and putative RNA-RNA interaction sites. Our data also revealed a core RNA-complex of smaller segments which serves as the foundation ('anchor') for the assembly of a complete network composed of ten ssRNA segments. The same order of core RNA complex formation was identified in cells transfected with viral RNAs. No viral protein was required for these assembly reactions. Further, substitution mutations in the interacting bases within the core assemblies, altered subsequent segment addition and affected virus replication. These data identify a wholly RNA driven reaction that may offer novel opportunities for designed attenuation or antiviral therapeutics.

2.
Viruses ; 16(4)2024 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-38675835

RESUMO

Many protein expression systems are primarily utilised to produce a single, specific recombinant protein. In contrast, most biological processes such as virus assembly rely upon a complex of several interacting proteins rather than the activity of a sole protein. The high complexity of the baculovirus genome, coupled with a multiphase replication cycle incorporating distinct transcriptional steps, made it the ideal system to manipulate for high-level expression of a single, or co-expression of multiple, foreign proteins within a single cell. We have developed and utilised a series of recombinant baculovirus systems to unravel the sequential assembly process of a complex non-enveloped model virus, bluetongue virus (BTV). The high protein yields expressed by the baculovirus system not only facilitated structure-function analysis of each viral protein but were also advantageous to crystallography studies and supported the first atomic-level resolution of a recombinant viral protein, the major BTV capsid protein. Further, the formation of recombinant double-shelled virus-like particles (VLPs) provided insights into the structure-function relationships among the four major structural proteins of the BTV whilst also representing a potential candidate for a viral vaccine. The baculovirus multi-gene expression system facilitated the study of structurally complex viruses (both non-enveloped and enveloped viruses) and heralded a new generation of viral vaccines.


Assuntos
Baculoviridae , Proteínas Recombinantes , Baculoviridae/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Animais , Expressão Gênica , Vírus Bluetongue/genética , Vetores Genéticos/genética , Montagem de Vírus , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/química
3.
Cell ; 187(9): 2236-2249.e17, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38614100

RESUMO

Unlike those of double-stranded DNA (dsDNA), single-stranded DNA (ssDNA), and ssRNA viruses, the mechanism of genome packaging of dsRNA viruses is poorly understood. Here, we combined the techniques of high-resolution cryoelectron microscopy (cryo-EM), cellular cryoelectron tomography (cryo-ET), and structure-guided mutagenesis to investigate genome packaging and capsid assembly of bluetongue virus (BTV), a member of the Reoviridae family of dsRNA viruses. A total of eleven assembly states of BTV capsid were captured, with resolutions up to 2.8 Å, with most visualized in the host cytoplasm. ATPase VP6 was found underneath the vertices of capsid shell protein VP3 as an RNA-harboring pentamer, facilitating RNA packaging. RNA packaging expands the VP3 shell, which then engages middle- and outer-layer proteins to generate infectious virions. These revealed "duality" characteristics of the BTV assembly mechanism reconcile previous contradictory co-assembly and core-filling models and provide insights into the mysterious RNA packaging and capsid assembly of Reoviridae members and beyond.


Assuntos
Vírus Bluetongue , Proteínas do Capsídeo , Capsídeo , Microscopia Crioeletrônica , RNA Viral , Empacotamento do Genoma Viral , Vírus Bluetongue/genética , Vírus Bluetongue/fisiologia , Vírus Bluetongue/metabolismo , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/química , Animais , RNA Viral/metabolismo , RNA Viral/genética , Genoma Viral/genética , Montagem de Vírus , Tomografia com Microscopia Eletrônica , Vírion/metabolismo , Vírion/genética , Vírion/ultraestrutura , Modelos Moleculares , Linhagem Celular , Cricetinae
4.
Nucleic Acids Res ; 51(10): 5210-5227, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37070191

RESUMO

How multi-segmented double-stranded RNA (dsRNA) viruses correctly incorporate their genomes into their capsids remains unclear for many viruses, including Bluetongue virus (BTV), a Reoviridae member, with a genome of 10 segments. To address this, we used an RNA-cross-linking and peptide-fingerprinting assay (RCAP) to identify RNA binding sites of the inner capsid protein VP3, the viral polymerase VP1 and the capping enzyme VP4. Using a combination of mutagenesis, reverse genetics, recombinant proteins and in vitro assembly, we validated the importance of these regions in virus infectivity. Further, to identify which RNA segments and sequences interact with these proteins, we used viral photo-activatable ribonucleoside crosslinking (vPAR-CL) which revealed that the larger RNA segments (S1-S4) and the smallest segment (S10) have more interactions with viral proteins than the other smaller segments. Additionally, using a sequence enrichment analysis we identified an RNA motif of nine bases that is shared by the larger segments. The importance of this motif for virus replication was confirmed by mutagenesis followed by virus recovery. We further demonstrated that these approaches could be applied to a related Reoviridae member, rotavirus (RV), which has human epidemic impact, offering the possibility of novel intervention strategies for a human pathogen.


Assuntos
Vírus Bluetongue , Capsídeo , RNA Viral , Proteínas Virais , Animais , Humanos , Vírus Bluetongue/química , Vírus Bluetongue/metabolismo , Capsídeo/química , Capsídeo/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Replicação Viral , Proteínas Virais/química , Proteínas Virais/metabolismo
5.
Structure ; 2023 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-36841235

RESUMO

The definition of structure as the arrangement of and relations between the parts of something complex has always been a challenge in virology. The balance required for a virus to be sufficiently stable to allow transmission yet also be primed for disassembly on contact with a permissive cell is precarious and seemingly difficult to attain. Add to this that virus structural components often have multiple functions such as receptor binding, fusion, and cleavage, and the puzzle deepens. It also has consequences: virus yields may be compromised, vaccine shelf-life may be limited, and the ability to quickly evolve away from an intervention may be underestimated. Progress in understanding virus structure and the ways in which it might be exploited were the subject of the latest International Virus Assembly Symposium. Whole viruses, individual components, and transient intermediates were revealed at sufficiently high resolution to deduce the mechanisms concerned.

6.
J Med Virol ; 95(1): e28252, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36271727

RESUMO

Zika virus (ZIKV) is one of several examples of an unprecedented pandemic spread and against which there is currently no suitable vaccine or treatment. Here, we constructed and characterized recombinant baculovirus-derived ZIKV-like particles (Zika VLPs) to study ZIKV-antibody interactions. These VLPs, uniquely consisted of the full-length ZIKV capsid (C), pre-membrane (prM), and envelope (E) proteins with either: a) the viral nonstructural NS2B and NS3 protease unit under one or two different promoters or b) an alternative host-cell furin protease encoding cleavage sequence inserted between the C and prM genes, together with lobster tropomyosin leader and honeybee signal sequences with one promoter for increased extracellular secretion. All these Zika VLPs displayed typical virion morphology in transmission electron microscopic analysis when expressed in both insect (Sf9) and mammalian (HEK293T) cells and no uncleaved prM glycoprotein was detected, as are present on immature virions. The importance of glycosylation of the E glycoprotein was shown by the effects on both polyclonal and monoclonal antibody reactions after these N-linked carbohydrate residues were disrupted by oxidation or enzymatic cleavage. Importantly, the construct which contained the host-cell furin protease cleavage sequence together with a lobster tropomyosin leader and honeybee signal sequences under one promoter produced higher Zika VLP titers and protein concentrations and which can now be tested as a superior construct in multifunctional diagnostic (ELISA and neutralization/antibody-dependent enhancement) assays and immunogenic assessments possibly leading to vaccine trials.


Assuntos
Infecção por Zika virus , Zika virus , Humanos , Animais , Infecção por Zika virus/prevenção & controle , Furina/metabolismo , Baculoviridae/genética , Células HEK293 , Tropomiosina/metabolismo , Sinais Direcionadores de Proteínas , Proteínas do Envelope Viral/genética , Mamíferos/metabolismo
7.
Nucleic Acids Res ; 50(19): 11273-11284, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36259663

RESUMO

Liquid-liquid phase separation (LLPS) has assumed a prominent role in biological cell systems, where it underpins the formation of subcellular compartments necessary for cell function. We investigated the underlying mechanism of LLPS in virus infected cells, where virus inclusion bodies are formed by an RNA-binding phosphoprotein (NS2) of Bluetongue virus to serve as sites for subviral particle assembly and virus maturation. We show that NS2 undergoes LLPS that is dependent on protein phosphorylation and RNA-binding and that LLPS occurrence is accompanied by a change in protein secondary structure. Site-directed mutagenesis identified two critical arginine residues in NS2 responsible for specific RNA binding and thus for NS2-RNA complex driven LLPS. Reverse genetics identified the same residues as essential for VIB assembly in infected cells and virus viability. Our findings suggest that a specific arginine-RNA interaction in the context of a phosphorylated state drives LLPS in this, and possibly other, virus infections.


Assuntos
Vírus Bluetongue , Montagem de Vírus , Animais , Fosforilação , Montagem de Vírus/genética , Vírus Bluetongue/genética , RNA/metabolismo , Arginina/metabolismo
8.
Viruses ; 14(5)2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35632656

RESUMO

Coronavirus Disease 2019 (COVID-19), caused by infection with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has highlighted the need for the rapid generation of efficient vaccines for emerging disease. Virus-like particles, VLPs, are an established vaccine technology that produces virus-like mimics, based on expression of the structural proteins of a target virus. SARS-CoV-2 is a coronavirus where the basis of VLP formation has been shown to be the co-expression of the spike, membrane and envelope structural proteins. Here we describe the generation of SARS-CoV-2 VLPs by the co-expression of the salient structural proteins in insect cells using the established baculovirus expression system. VLPs were heterologous ~100 nm diameter enveloped particles with a distinct fringe that reacted strongly with SARS-CoV-2 convalescent sera. In a Syrian hamster challenge model, non-adjuvanted VLPs induced neutralizing antibodies to the VLP-associated Wuhan S protein and reduced virus shedding and protected against disease associated weight loss following a virulent challenge with SARS-CoV-2 (B.1.1.7 variant). Immunized animals showed reduced lung pathology and lower challenge virus replication than the non-immunized controls. Our data suggest SARS-CoV-2 VLPs offer an efficient vaccine that mitigates against virus load and prevents severe disease.


Assuntos
Baculoviridae , COVID-19 , Animais , Baculoviridae/genética , COVID-19/prevenção & controle , COVID-19/terapia , Cricetinae , Humanos , Imunização Passiva , SARS-CoV-2/genética , Soroterapia para COVID-19
9.
J Virol ; 96(1): e0167721, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34669428

RESUMO

Bluetongue virus (BTV), a member of the Orbivirus genus, is transmitted by biting midges (gnats, Culicoides sp.) and is one of the most widespread animal pathogens, causing serious outbreaks in domestic animals, particularly in sheep, with high economic impact. The non-enveloped BTV particle is a double-capsid structure of seven proteins and a genome of 10 double-stranded RNA segments. Although the outermost spike-like VP2 acts as the attachment protein during BTV entry, no specific host receptor has been identified for BTV. Recent high-resolution cryo-electron (cryoEM) structures and biological data have suggested that VP2 may interact with sialic acids (SAs). To confirm this, we have generated protein-based nanoparticles displaying multivalent VP2 and used them to probe glycan arrays. The data show that VP2 binds α2,3-linked SA with high affinity but also binds α2,6-linked SA. Further, Maackia amurensis lectin II (MAL II) and Sambucus nigra lectin (SNA), which specifically bind α2,3-linked and α2,6-linked SAs, respectively, inhibited BTV infection and virus growth in susceptible sheep cells while SNA alone inhibited virus growth in Culicoides-derived cells. A combination of hydrogen deuterium exchange mass spectrometry and site-directed mutagenesis allowed the identification of the specific SA binding residues of VP2. This study provides direct evidence that sialic acids act as key receptor for BTV and that the outer capsid protein VP2 specifically binds SA during BTV entry in both mammalian and insect cells. IMPORTANCE To date no receptor has been assigned for non-enveloped bluetongue virus. To determine if the outermost spike-like VP2 protein is responsible for host cell attachment via interaction with sialic acids, we first generated a protein-based VP2-nanoparticle, for the multivalent presentation of recombinant VP2 protein. Using nanoparticles displaying VP2 to probe a glycan array, we identified that VP2 binds both α2,3-linked and α2,6-linked sialic acids. Lectin inhibitors targeting both linkages of sialic acids showed strong inhibition to BTV infection and progeny virus production in mammalian cells; however the inhibition was only seen with the lectin targeting α2,6-linked sialic acid in insect vector cells. In addition, we identified the VP2 sialic acid binding sites in the exposed tip domain. Our data provides direct evidence that sialic acids act as key receptors for BTV attachment and entry in to both mammalian and insect cells.


Assuntos
Sítios de Ligação , Vírus Bluetongue/fisiologia , Bluetongue/virologia , Proteínas do Capsídeo/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Interações Hospedeiro-Patógeno , Lectinas/metabolismo , Espectrometria de Massas , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Receptores Virais/química , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo
10.
Nat Microbiol ; 6(11): 1424-1432, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34702979

RESUMO

Bluetongue virus (BTV) is a non-enveloped virus and causes substantial morbidity and mortality in ruminants such as sheep. Fashioning a receptor-binding protein (VP2) and a membrane penetration protein (VP5) on the surface, BTV releases its genome-containing core (VP3 and VP7) into the host cell cytosol after perforation of the endosomal membrane. Unlike enveloped ones, the entry mechanisms of non-enveloped viruses into host cells remain poorly understood. Here we applied single-particle cryo-electron microscopy, cryo-electron tomography and structure-guided functional assays to characterize intermediate states of BTV cell entry in endosomes. Four structures of BTV at the resolution range of 3.4-3.9 Å show the different stages of structural rearrangement of capsid proteins on exposure to low pH, including conformational changes of VP5, stepwise detachment of VP2 and a small shift of VP7. In detail, sensing of the low-pH condition by the VP5 anchor domain triggers three major VP5 actions: projecting the hidden dagger domain, converting a surface loop to a protonated ß-hairpin that anchors VP5 to the core and stepwise refolding of the unfurling domains into a six-helix stalk. Cryo-electron tomography structures of BTV interacting with liposomes show a length decrease of the VP5 stalk from 19.5 to 15.5 nm after its insertion into the membrane. Our structures, functional assays and structure-guided mutagenesis experiments combined indicate that this stalk, along with dagger domain and the WHXL motif, creates a single pore through the endosomal membrane that enables the viral core to enter the cytosol. Our study unveils the detailed mechanisms of BTV membrane penetration and showcases general methods to study cell entry of other non-enveloped viruses.


Assuntos
Vírus Bluetongue/metabolismo , Bluetongue/virologia , Proteínas do Capsídeo/metabolismo , Endossomos/virologia , Animais , Vírus Bluetongue/química , Vírus Bluetongue/genética , Vírus Bluetongue/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Endossomos/química , Concentração de Íons de Hidrogênio , Modelos Moleculares , Ovinos , Doenças dos Ovinos/virologia , Internalização do Vírus
11.
Viruses ; 13(9)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34578422

RESUMO

Understanding how viruses with multi-segmented genomes incorporate one copy of each segment into their capsids remains an intriguing question. Here, we review our recent progress and describe the advancements made in understanding the genome packaging mechanism of a model nonenveloped virus, Bluetongue virus (BTV), with a 10-segment (S1-S10) double-strand RNA (dsRNA) genome. BTV (multiple serotypes), a member of the Orbivirus genus in the Reoviridae family, is a notable pathogen for livestock and is responsible for significant economic losses worldwide. This has enabled the creation of an extensive set of reagents and assays, including reverse genetics, cell-free RNA packaging, and bespoke bioinformatics approaches, which can be directed to address the packaging question. Our studies have shown that (i) UTRs enable the conformation of each segment necessary for the next level of RNA-RNA interaction; (ii) a specific order of intersegment interactions leads to a complex RNA network containing all the active components in sorting and packaging; (iii) networked segments are recruited into nascent assembling capsids; and (iv) select capsid proteins might be involved in the packaging process. The key features of genome packaging mechanisms for BTV and related dsRNA viruses are novel and open up new avenues of potential intervention.


Assuntos
Vírus Bluetongue/genética , Vírus Bluetongue/fisiologia , RNA Viral/metabolismo , Empacotamento do Genoma Viral , Montagem de Vírus , Replicação Viral , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Genoma Viral , Conformação de Ácido Nucleico , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/metabolismo , RNA Viral/química , Proteínas não Estruturais Virais/metabolismo
12.
Vaccine ; 39(23): 3161-3168, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33958224

RESUMO

African horse sickness virus (AHSV) is an Orbivirus within the Reoviridae family, spread by Culicoides species of midges, which infects equids with high mortality, particularly in horses and has a considerable impact on the equine industry. In order to control the disease, we previously described Entry Competent Replication Abortive (ECRA) virus strains for each of the nine distinct AHSV serotypes and demonstrated their potential as vaccines, first in type I interferon receptor (IFNAR-/-) knockout mice, and then in ponies. In this report we have investigated whether or not a combination ECRA vaccine comprising nine vaccine strains as two different cocktails is as efficient in ponies and the duration of the immunity triggered by ECRA vaccines. In one study, a group of ponies were vaccinated with a cocktail of 4 vaccine strains, followed by a vaccination of the remaining 5 vaccine strains, mimicking the current live attenuated vaccine regimen. In the second study, ponies were vaccinated with a single ECRA-AHSV strain and monitored for 6 months. The first group of ponies developed neutralising antibody responses against all 9 serotypes, indicating that no cross-serotype interference occurred, while the second group developed robust neutralising antibody responses against the single serotype that were sustained at the same level throughout a 6-month study. The results support our previous data and further validate ECRA vaccines as a safe and efficacious replacement of current live vaccines.


Assuntos
Vírus da Doença Equina Africana , Doença Equina Africana , Vacinas Virais , Doença Equina Africana/prevenção & controle , Vírus da Doença Equina Africana/genética , Animais , Cavalos , Camundongos , Sorogrupo , Vacinas Atenuadas
13.
Lancet ; 397(10275): 642-643, 2021 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-33545098
14.
PLoS Pathog ; 16(10): e1009015, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33075107

RESUMO

Recent developments on extracellular vesicles (EVs) containing multiple virus particles challenge the rigid definition of non-enveloped viruses. However, how non-enveloped viruses hijack cell machinery to promote non-lytic release in EVs, and their functional roles, remain to be clarified. Here we used Bluetongue virus (BTV) as a model of a non-enveloped arthropod-borne virus and discovered that the majority of viruses are released in EVs. Based on the cellular proteins detected in these EVs, and use of inhibitors targeting the cellular degradation process, we demonstrated that these extracellular vesicles are derived from secretory lysosomes, in which the acidic pH is neutralized upon the infection. Moreover, we report that secreted EVs are more efficient than free-viruses for initiating infections, but that they trigger super-infection exclusion that only free-viruses can overcome.


Assuntos
Vírus Bluetongue/metabolismo , Vesículas Extracelulares/metabolismo , Lisossomos/metabolismo , Animais , Arbovírus/patogenicidade , Vírus Bluetongue/genética , Linhagem Celular , Vesículas Extracelulares/fisiologia , Modelos Biológicos , Vesículas Secretórias , Vírion , Replicação Viral
15.
J Virol ; 94(20)2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32759321

RESUMO

Many viruses use specific viral proteins to bind calcium ions (Ca2+) for stability or to modify host cell pathways; however, to date, no Ca2+ binding protein has been reported in bluetongue virus (BTV), the causative agent of bluetongue disease in livestock. Here, using a comprehensive bioinformatics screening, we identified a putative EF-hand-like Ca2+ binding motif in the carboxyl terminal region of BTV nonstructural phosphoprotein 2 (NS2). Subsequently, using a recombinant NS2, we demonstrated that NS2 binds Ca2+ efficiently and that Ca2+ binding was perturbed when the Asp and Glu residues in the motif were substituted by alanine. Using circular dichroism analysis, we found that Ca2+ binding by NS2 triggered a helix-to-coil secondary structure transition. Further, cryo-electron microscopy in the presence of Ca2+ revealed that NS2 forms helical oligomers which, when aligned with the N-terminal domain crystal structure, suggest an N-terminal domain that wraps around the C-terminal domain in the oligomer. Further, an in vitro kinase assay demonstrated that Ca2+ enhanced the phosphorylation of NS2 significantly. Importantly, mutations introduced at the Ca2+ binding site in the viral genome by reverse genetics failed to allow recovery of viable virus, and the NS2 phosphorylation level and assembly of viral inclusion bodies (VIBs) were reduced. Together, our data suggest that NS2 is a dedicated Ca2+ binding protein and that calcium sensing acts as a trigger for VIB assembly, which in turn facilitates virus replication and assembly.IMPORTANCE After entering the host cells, viruses use cellular host factors to ensure a successful virus replication process. For replication in infected cells, members of the Reoviridae family form inclusion body-like structures known as viral inclusion bodies (VIB) or viral factories. Bluetongue virus (BTV) forms VIBs in infected cells through nonstructural protein 2 (NS2), a phosphoprotein. An important regulatory factor critical for VIB formation is phosphorylation of NS2. In our study, we discovered a characteristic calcium-binding EF-hand-like motif in NS2 and found that the calcium binding preferentially affects phosphorylation level of the NS2 and has a role in regulating VIB assembly.


Assuntos
Vírus Bluetongue/química , Vírus Bluetongue/fisiologia , Cálcio/química , Proteínas não Estruturais Virais/química , Replicação Viral , Animais , Sítios de Ligação , Cálcio/metabolismo , Linhagem Celular , Dicroísmo Circular , Cricetinae , Cristalografia por Raios X , Estrutura Secundária de Proteína , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
16.
Curr Opin Virol ; 44: 35-41, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32610251

RESUMO

Bluetongue virus (BTV) reverse genetics (RG), available since 2007, has allowed the dissection of the virus replication cycle, including discovery of a primary replication stage. This information has allowed the generation of Entry-Competent-Replication-Abortive (ECRA) vaccines, which enter cells and complete primary replication but fail to complete the later stage. A series of vaccine trials in sheep and cattle either with a single ECRA serotype or a cocktail of multiple ECRA serotypes have demonstrated that these vaccines provide complete protection against virulent virus challenge without cross-serotype interference. Similarly, an RG system developed for the related African Horse Sickness virus, which causes high mortality in equids has provided AHSV ECRA vaccines that are protective in horses. ECRA vaccines were incapable of productive replication in animals despite being competent for cell entry. This technology allows rapid generation of emerging Orbivirus vaccines and offers immunogenicity and safety levels that surpass attenuated or recombinant routes.


Assuntos
Vírus Bluetongue/genética , Vírus Bluetongue/imunologia , Orbivirus/genética , Orbivirus/imunologia , Infecções por Reoviridae/veterinária , Genética Reversa/métodos , Vacinas Virais/genética , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/imunologia , Bluetongue/imunologia , Bluetongue/prevenção & controle , Bovinos , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/prevenção & controle , Ovinos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Replicação Viral
17.
Microorganisms ; 8(7)2020 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-32605099

RESUMO

Bluetongue virus (BTV) is an arthropod-borne virus infecting livestock. Its frequent emergence in Europe and North America had caused significant agricultural and economic loss. BTV is also of scientific interest as a model to understand the mechanisms underlying non-enveloped virus release from mammalian and insect cells. The BTV particle, which is formed of a complex double-layered capsid, was first considered as a lytic virus that needs to lyse the infected cells for cell to cell transmission. In the last decade, however, a more in-depth focus on the role of the non-structural proteins has led to several examples where BTV particles are also released through different budding mechanisms at the plasma membrane. It is now clear that the non-structural protein NS3 is the main driver of BTV release, via different interactions with both viral and cellular proteins of the cell sorting and exocytosis pathway. In this review, we discuss the most recent advances in the molecular biology of BTV egress and compare the mechanisms that lead to lytic or non-lytic BTV release.

18.
Viruses ; 12(3)2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32245145

RESUMO

Members of the Reoviridae family assemble virus factories within the cytoplasm of infected cells to replicate and assemble virus particles. Bluetongue virus (BTV) forms virus inclusion bodies (VIBs) that are aggregates of viral RNA, certain viral proteins, and host factors, and have been shown to be sites of the initial assembly of transcriptionally active virus-like particles. This study sought to characterize the formation, composition, and ultrastructure of VIBs, particularly in relation to virus replication. In this study we have utilized various microscopic techniques, including structured illumination microscopy, and virological assays to show for the first time that the outer capsid protein VP5, which is essential for virus maturation, is also associated with VIBs. The addition of VP5 to assembled virus cores exiting VIBs is required to arrest transcriptionally active core particles, facilitating virus maturation. Furthermore, we observed a time-dependent association of the glycosylated non-structural protein 3 (NS3) with VIBs, and report on the importance of the two polybasic motifs within NS3 that facilitate virus trafficking and egress from infected cells at the plasma membrane. Thus, the presence of VP5 and the dynamic nature of NS3 association with VIBs that we report here provide novel insight into these previously less well-characterized processes.


Assuntos
Vírus Bluetongue/fisiologia , Proteínas não Estruturais Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Replicação Viral , Animais , Proteínas do Capsídeo , Linhagem Celular , Cobaias , Camundongos , Ligação Proteica , Transporte Proteico , Coelhos , Proteínas não Estruturais Virais/genética , Proteínas Estruturais Virais/genética
19.
Adv Virus Res ; 108: 249-273, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33837718

RESUMO

Bluetongue virus (BTV) is an insect-vectored emerging pathogen of wild ruminants and livestock in many parts of the world. The virion particle is a complex structure of consecutive layers of protein surrounding a genome of 10 double-stranded (ds) RNA segments. BTV has been studied extensively as a model system for large, nonenveloped dsRNA viruses. A combination of recombinant proteins and particles together with reverse genetics, high-resolution structural analysis by X-ray crystallography and cryo-electron microscopy techniques have been utilized to provide an order for the assembly of the capsid shell and the protein sequestration required for it. Further, a reconstituted in vitro assembly system and RNA-RNA interaction assay, have defined the individual steps required for the assembly and packaging of the 10-segmented RNA genome. In addition, various microscopic techniques have been utilized to illuminate the stages of virus maturation and its egress via multiple pathways. These findings have not only given an overall understanding of BTV assembly and morphogenesis but also indicated that similar assembly and egress pathways are likely to be used by related viruses and provided an informed starting point for intervention or prevention.


Assuntos
Vírus Bluetongue/fisiologia , Capsídeo/fisiologia , Montagem de Vírus , Liberação de Vírus , Animais , Vírus Bluetongue/genética , Vírus Bluetongue/patogenicidade , Vírus Bluetongue/ultraestrutura , Livros , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Genoma Viral , Insetos Vetores/virologia , Gado/virologia , Proteínas Virais/genética , Vírion/metabolismo , Vírion/patogenicidade , Replicação Viral
20.
Viruses ; 11(12)2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31795485

RESUMO

Bluetongue virus (BTV) is an arthropod-borne virus that infects domestic and wild ruminants. The virion is a non-enveloped double-layered particle with an outer capsid that encloses a core containing the segmented double-stranded RNA genome. Although BTV is canonically released by cell lysis, it also exits non-lytically. In infected cells, the BTV nonstructural glycoprotein 3 (NS3) is found to be associated with host membranes and traffics from the endoplasmic reticulum through the Golgi apparatus to the plasma membrane. This suggests a role for NS3 in BTV particle maturation and non-lytic egress. However, the mechanism by which NS3 coordinates these events has not yet been elucidated. Here, we identified two polybasic motifs (PMB1/PMB2), consistent with the membrane binding. Using site-directed mutagenesis, confocal and electron microscopy, and flow cytometry, we demonstrated that PBM1 and PBM2 mutant viruses retained NS3 either in the Golgi apparatus or in the endoplasmic reticulum, suggesting a distinct role for each motif. Mutation of PBM2 motif decreased NS3 export to the cell surface and virus production. However, both mutant viruses produced predominantly inner core particles that remained close to their site of assembly. Together, our data demonstrates that correct trafficking of the NS3 protein is required for virus maturation and release.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/virologia , Proteínas não Estruturais Virais/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Vírus Bluetongue/genética , Vírus Bluetongue/ultraestrutura , Cricetinae , Mutagênese Sítio-Dirigida , Transporte Proteico , Alinhamento de Sequência , Ovinos , Proteínas não Estruturais Virais/genética , Vírion , Montagem de Vírus , Liberação de Vírus , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA