Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Vaccine ; 38(4): 882-889, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31708178

ABSTRACT

African horse sickness virus (AHSV) is an insect-borne pathogen that causes acute disease in horses and other equids. In an effort to improve the safety of currently available vaccines and to acquire new knowledge about the determinants of AHSV immunogenicity, new generation vaccines are being developed. In this work we have generated and tested a novel immunization approach comprised of nonstructural protein 1 (NS1) of AHSV serotype 4 (AHSV-4) incorporated into avian reovirus muNS protein microspheres (MS-NS1) and/or expressed using recombinant modified vaccinia virus Ankara vector (MVA-NS1). The protection conferred against AHSV by a homologous MS-NS1 or heterologous MS-NS1 and MVA-NS1 prime/boost was evaluated in IFNAR (-/-) mice. Our results indicate that immunization based on MS-NS1 and MVA-NS1 afforded complete protection against the infection with homologous AHSV-4. Moreover, priming with MS-NS1 and boost vaccination with MVA-NS1 (MS-MVA-NS1) triggered NS1 specific cytotoxic CD8 + T cells and prevented AHSV disease in IFNAR (-/-) mice after challenge with heterologous serotype AHSV-9. Cross-protective immune responses are highly important since AHS can be caused by nine different serotypes, which means that a universal polyvalent vaccination would need to induce protective immunity against all serotypes.


Subject(s)
African Horse Sickness Virus/immunology , African Horse Sickness/prevention & control , Immunization , Viral Vaccines/administration & dosage , African Horse Sickness/immunology , Animals , Female , Horses , Immunity/immunology , Mice , Mice, Knockout , Microspheres , Orthoreovirus, Avian/immunology , Receptor, Interferon alpha-beta/genetics , Serogroup , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology
2.
Sci Rep ; 8(1): 16286, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30390011

ABSTRACT

We have previously developed a methodology to produce protein microspheres (MS) that can be loaded with proteins of interest in living cells through their C or N-terminal tagging with the so-called IC-Tag. The IC-Tagging method has many applications ranging from the production of immobilized enzymes for industrial use to the production of subunit vaccines due to its intrinsic adjuvancy. Here we show the adaptation of the IC-Tagging to work inside the endoplasmic reticulum and bacteria, allowing us to produce properly modified viral glycoproteins. Additionally, we were able to express the Islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), whose expression remained elusive to date possibly due to its toxicity when over-expressed. IGRP is an antigen of enormous pharmaceutical interest as it is specifically targeted during the autoimmune response taking place in both the Non-Obese Diabetic (NOD) mice and type 1 diabetes (T1D) patients leading to the destruction of insulin-producing beta cells.


Subject(s)
Autoantigens/isolation & purification , Glucose-6-Phosphatase/isolation & purification , Inclusion Bodies, Viral/metabolism , Recombinant Fusion Proteins/isolation & purification , Animals , Autoantigens/genetics , Autoantigens/metabolism , Cell Line , Chick Embryo , Genetic Vectors/genetics , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycoproteins/genetics , Orthoreovirus, Avian/genetics , Plasmids/genetics , Protein Domains/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification , Viral Nonstructural Proteins/metabolism
3.
Antiviral Res ; 142: 55-62, 2017 06.
Article in English | MEDLINE | ID: mdl-28322923

ABSTRACT

Bluetongue virus (BTV) is the causative agent of bluetongue disease (BT), which affects domestic and wild ruminants. At the present, 27 different serotypes have been documented. Vaccination has been demonstrated as one of the most effective methods to avoid viral dissemination. To overcome the drawbacks associated with the use of inactivated and attenuated vaccines we engineered a new recombinant BTV vaccine candidate based on proteins VP2, VP7, and NS1 of BTV-4 that were incorporated into avian reovirus muNS-Mi microspheres (MS-VP2/VP7/NS1) and recombinant modified vaccinia virus Ankara (rMVA). The combination of these two antigen delivery systems in a heterologous prime-boost vaccination strategy generated significant levels of neutralizing antibodies in IFNAR(-/-) mice. Furthermore, this immunization strategy increased the ratio of IgG2a/IgG1 in sera, indicating an induction of a Th1 response, and elicited a CD8 T cell response. Immunized mice were protected against lethal challenges with the homologous serotype 4 and the heterologous serotype 1 of BTV. All these results support the strategy based on microspheres in combination with rMVAs as a promising multiserotype vaccine candidate against BTV.


Subject(s)
Bluetongue virus/immunology , Bluetongue/prevention & control , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Microspheres , Receptor, Interferon alpha-beta/immunology , Vaccination , Vaccinia virus/immunology , Animals , Antibodies, Neutralizing , Bluetongue virus/classification , CD8-Positive T-Lymphocytes/metabolism , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Line , Chlorocebus aethiops , Immunization , Immunization, Secondary/methods , Immunoglobulin G/blood , Lysosomal-Associated Membrane Protein 1/metabolism , Male , Mice , Mice, 129 Strain , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/immunology , Serogroup , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology , Vero Cells , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology
4.
J Virol ; 90(18): 8328-40, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27440902

ABSTRACT

UNLABELLED: We have previously shown that the replication of avian reovirus (ARV) in chicken cells is much more resistant to interferon (IFN) than the replication of vesicular stomatitis virus (VSV) or vaccinia virus (VV). In this study, we have investigated the role that the double-stranded RNA (dsRNA)-activated protein kinase (PKR) plays in the sensitivity of these three viruses toward the antiviral action of chicken interferon. Our data suggest that while interferon priming of avian cells blocks vaccinia virus replication by promoting PKR activation, the replication of vesicular stomatitis virus appears to be blocked at a pretranslational step. Our data further suggest that the replication of avian reovirus in chicken cells is quite resistant to interferon priming because this virus uses strategies to downregulate PKR activation and also because translation of avian reovirus mRNAs is more resistant to phosphorylation of the alpha subunit of initiation factor eIF2 than translation of their cellular counterparts. Our results further reveal that the avian reovirus protein sigmaA is able to prevent PKR activation and that this function is dependent on its double-stranded RNA-binding activity. Finally, this study demonstrates that vaccinia virus and avian reovirus, but not vesicular stomatitis virus, express/induce factors that counteract the ability of dithiothreitol to promote eIF2 phosphorylation. Our data demonstrate that each of the three different viruses used in this study elicits distinct responses to interferon and to dithiothreitol-induced eIF2 phosphorylation when infecting avian cells. IMPORTANCE: Type I interferons constitute the first barrier of defense against viral infections, and one of the best characterized antiviral strategies is mediated by the double-stranded RNA-activated protein kinase R (PKR). The results of this study revealed that IFN priming of avian cells has little effect on avian reovirus (ARV) replication but drastically diminishes the replication of vaccinia virus (VV) and vesicular stomatitis virus (VSV) by PKR-dependent and -independent mechanisms, respectively. Our data also demonstrate that the dsRNA-binding ability of ARV protein sigmaA plays a key role in the resistance of ARV toward IFN by preventing PKR activation. Our findings will contribute to improve the current understanding of the interaction of viruses with the host's innate immune system. Finally, it would be of interest to uncover the mechanisms that allow avian reovirus transcripts to be efficiently translated under conditions (moderate eIF2 phosphorylation) that block the synthesis of cellular proteins.


Subject(s)
Antiviral Agents/metabolism , Interferons/metabolism , Orthoreovirus, Avian/physiology , Vaccinia virus/physiology , Vesiculovirus/physiology , Virus Replication/drug effects , eIF-2 Kinase/metabolism , Animals , Cell Line , Chickens , Dithiothreitol/metabolism , Eukaryotic Initiation Factor-2/metabolism , Host-Pathogen Interactions , Phosphorylation , Protein Processing, Post-Translational
5.
Methods Mol Biol ; 1349: 25-34, 2016.
Article in English | MEDLINE | ID: mdl-26458827

ABSTRACT

Particulate material is more efficient in eliciting immune responses. Here we describe the production of microspheres formed by protein muNS-Mi from avian reoviruses, loaded with foreign epitopes by means of IC-Tagging, for their use as vaccines.


Subject(s)
Epitopes/immunology , Orthoreovirus, Avian/drug effects , Vaccination/methods , Viral Nonstructural Proteins/immunology , Animals , Birds/immunology , Birds/virology , Fluorescence , Microspheres , Orthoreovirus, Avian/pathogenicity
6.
Virology ; 487: 104-11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517397

ABSTRACT

We have previously shown that the replication of avian reovirus (ARV) in chicken embryo fibroblasts (CEF) is more resistant to the antiviral action of interferon (IFN) than the replication of vesicular stomatitis virus (VSV) or vaccinia virus (VV). In this study we examined the capacity of these three viruses to induce the expression of IFN when infecting avian cells. Efficient expression of both type-α and type-ß IFNs, as well as of the double-stranded RNA (dsRNA)-activated protein kinase (PKR), takes place in ARV-infected CEF, but not in cells infected with VSV or VV. PKR expression is not directly induced by ARV infection, but by the IFN secreted by ARV-infected cells. IFN induction in ARV-infected cells requires viral uncoating, but not viral gene expression, a situation similar to that reported for apoptosis induction by ARV-infected cells. However, our results demonstrate that IFN induction by ARV-infected CEF occurs by a caspase-independent mechanism.


Subject(s)
Interferon-alpha/immunology , Interferon-beta/immunology , Orthoreovirus, Avian/immunology , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/immunology , eIF-2 Kinase/immunology , Animals , Apoptosis/immunology , Caspases/metabolism , Cell Proliferation , Cells, Cultured , Chick Embryo , Chickens , Cricetinae , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Orthoreovirus, Avian/growth & development , Primary Cell Culture , Vaccinia virus/growth & development , Vesicular stomatitis Indiana virus/growth & development , Virus Replication , eIF-2 Kinase/biosynthesis
7.
Virology ; 462-463: 49-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25092461

ABSTRACT

Avian reovirus non-structural protein muNS is partially cleaved in infected chicken embryo fibroblast cells to produce a 55-kDa carboxyterminal protein, termed muNSC, and a 17-kDa aminoterminal polypeptide, designated muNSN. In this study we demonstrate that muNS processing is catalyzed by a caspase 3-like protease activated during the course of avian reovirus infection. The cleavage site was mapped by site directed mutagenesis between residues Asp-154 and Ala-155 of the muNS sequence. Although muNS and muNSC, but not muNSN, are able to form inclusions when expressed individually in transfected cells, only muNS is able to recruit specific ARV proteins to these structures. Furthermore, muNSC associates with ARV factories more weakly than muNS, sigmaNS and lambdaA. Finally, the inhibition of caspase activity in ARV-infected cells does not diminish ARV gene expression and replication, but drastically reduces muNS processing and the release and dissemination of progeny viral particles.


Subject(s)
Apoptosis , Caspase 3/metabolism , Host-Pathogen Interactions , Orthoreovirus, Avian/physiology , Protein Processing, Post-Translational , Viral Nonstructural Proteins/metabolism , Animals , Cells, Cultured , Chickens , Fibroblasts/virology
8.
Antiviral Res ; 110: 42-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25057758

ABSTRACT

Vaccination is critical for controlling the spread of bluetongue virus (BTV). The inactivated BTV vaccines that are now being used in Europe are effective in preventing outbreaks of BTV but secondary effects associated to repetitive inoculation of aluminum-containing adjuvants and the need to develop safer, cross-reactive, and more efficacious vaccines with differential diagnostic capability have re-stimulated the interest in developing improved vaccination strategies against BTV. We have engineered a subunit BTV vaccine candidate based on proteins VP2, VP7, and NS1 of BTV-4 incorporated into avian reovirus (ARV) muNS-Mi microspheres (MS-VP2/MS-VP7/MS-NS1). IFNAR(-/-) mice immunized with MS-VP2/MS-VP7/MS-NS1 without adjuvant generated significant levels of neutralizing antibodies specific to BTV-4. In addition, vaccination stimulated specific T cell responses, predominantly CD4+, against the virus. Immunized mice were fully protected against a homologous challenge with a lethal dose of BTV-4 and partially cross-protected against a heterologous challenge with a lethal dose of BTV-1. These results support MS-VP2/MS-VP7/MS-NS1 as a promising subunit vaccine candidate against multiple serotypes of BTV as well as the use of microspheres as an alternative delivery method with potent intrinsic adjuvant activity.


Subject(s)
Bluetongue virus/immunology , Capsid Proteins/immunology , Orthoreovirus, Avian/immunology , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Baculoviridae/genetics , Bluetongue/immunology , Bluetongue/prevention & control , CD4-Positive T-Lymphocytes/immunology , Capsid Proteins/genetics , Cell Line , Chick Embryo , Chlorocebus aethiops , Cricetinae , Drug Carriers/therapeutic use , Male , Mice , Mice, Knockout , Microspheres , Orthoreovirus, Avian/genetics , Sf9 Cells , Sheep/virology , Spodoptera , Vaccination , Vaccines, Subunit/immunology , Vero Cells , Viral Nonstructural Proteins/genetics
9.
Virology ; 432(2): 495-504, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22832119

ABSTRACT

A comparative analysis of the intracellular distribution of avian reovirus (ARV) core protein sigmaA in cells of avian and mammalian origin revealed that, whereas the viral protein accumulates in the cytoplasm and nucleolus of avian cells, most sigmaA concentrates in the nucleoplasm of mammalian cells in tight association with the insoluble nuclear matrix fraction. Our results further showed that sigmaA becomes arrested in the nucleoplasm of mammalian cells via association with mammalian cell-specific factors and that this association prevents nucleolar targeting. Inhibition of RNA polymerase II activity, but not of RNA polymerase I activity, in infected mammalian cells induces nucleus-to-cytoplasm sigmaA translocation through a CRM1- and RanGTP-dependent mechanism, yet a heterokaryon assay suggests that sigmaA does not shuttle between the nucleus and cytoplasm. The scarcity of sigmaA in cytoplasmic viral factories of infected mammalian cells could be one of the factors contributing to limited ARV replication in mammalian cells.


Subject(s)
Orthoreovirus, Avian/metabolism , RNA-Binding Proteins/metabolism , Subcellular Fractions/metabolism , Viral Core Proteins/metabolism , Animals , Birds/virology , Cell Line , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Chick Embryo/virology , Chlorocebus aethiops , Cytoplasm/metabolism , Fibroblasts/virology , HeLa Cells/virology , Humans , Mammals/virology , Species Specificity , Subcellular Fractions/virology , Vero Cells/virology
10.
J Biotechnol ; 155(3): 284-6, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21802457

ABSTRACT

We have recently developed a versatile tagging system (IC-tagging) that causes relocation of the tagged proteins to ARV muNS-derived intracellular globular inclusions. In the present study we demonstrate (i) that the IC-tag can be successfully fused either to the amino or carboxyl terminus of the protein to be tagged and (ii) that IC-tagged proteins are able to interact between them and perform complex reactions that require such interactions while integrated into muNS inclusions, increasing the versatility of the IC-tagging system. Also, our studies with the DsRed protein add some light on the structure/function relationship of the evolution of DsRed chromophore.


Subject(s)
Inclusion Bodies, Viral/metabolism , Molecular Probe Techniques , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , Immunohistochemistry , Inclusion Bodies, Viral/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Models, Molecular , Orthoreovirus, Avian/chemistry , Protein Binding , Protein Structure, Tertiary , Vero Cells , Viral Proteins/chemistry , Red Fluorescent Protein
11.
J Gen Virol ; 92(Pt 11): 2566-2574, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21795469

ABSTRACT

Previous reports revealed that the M3 gene of both avian and mammalian reoviruses express two isoforms of the non-structural protein µNS in infected cells. The larger isoforms initiate translation at the AUG codon closest to the 5' end of their respective m3 mRNAs, and were therefore designated µNS. In this study we have performed experiments to identify the molecular mechanisms by which the smaller µNS isoforms are generated. The results of this study confirmed the previous findings indicating that the smaller mammalian reovirus µNS isoform is a primary translation product, the translation of which is initiated at the internal AUG-41 codon of mammalian reovirus m3 mRNA. Our results further revealed that the smaller avian reovirus µNS isoform originates from a specific post-translational cleavage site near the amino terminus of µNS. This cleavage produces a 55 kDa carboxy-terminal protein, termed µNSC, and a 17 kDa amino-terminal polypeptide, designated µNSN. These results allowed us to extend the known avian reovirus protein-encoding capacity to 18 proteins, 12 of which are structural proteins and six of which are non-structural proteins. Our finding that avian and mammalian reoviruses use different mechanisms to express their µNSC isoforms suggests that these isoforms are important for reovirus replication.


Subject(s)
Orthoreovirus, Avian/physiology , Orthoreovirus, Mammalian/physiology , Protein Biosynthesis , Viral Nonstructural Proteins/biosynthesis , Animals , Cells, Cultured , Mice , Molecular Sequence Data , Protein Isoforms/biosynthesis , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Viral/genetics
12.
PLoS One ; 5(11): e13785, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-21072177

ABSTRACT

BACKGROUND: Characterization of protein-protein interactions is essential for understanding cellular functions. Although there are many published methods to analyze protein-protein interactions, most of them present serious limitations. In a different study we have characterized a novel avian reovirus muNS-based protein tagging and inclusion targeting method, and demonstrated its validity to purify free an immobilized protein. METHODOLOGY/PRINCIPAL FINDINGS: Here we present a method to identify protein-protein interactions inside living eukaryotic cells (tested in primate and avian cells). When p53 was tagged with Intercoil (IC; muNS residues 477-542), it not only got integrated into muNS cytoplasmic inclusions, but also attracted its known ligand SV40 large T antigen (TAg) to these structures. We have also adapted this system to work within the cell nucleus, by creating muNS-related protein chimeras that form nuclear inclusions. We show that nuclear muNS-derived inclusions are as efficient as cytoplasmic ones in capturing IC-tagged proteins, and that the proteins targeted to nuclear inclusions are able to interact with their known ligands. CONCLUSIONS/SIGNIFICANCE: Our protein redistribution method does not present the architectural requirement of re-constructing a transcription factor as any of the two-hybrid systems do. The method is simple and requires only cell transfection and a fluorescence microscope. Our tagging method can be used either in the cytoplasm or the nucleus of living cells to test protein-protein interactions or to perform functional studies by protein ligand sequestration.


Subject(s)
Inclusion Bodies, Viral/metabolism , Peptide Fragments/metabolism , Protein Interaction Mapping/methods , Tumor Suppressor Protein p53/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Cells, Cultured , Chick Embryo , Chlorocebus aethiops , Cytoplasm/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/metabolism , Peptide Fragments/genetics , Protein Binding , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
13.
PLoS One ; 5(11): e13961, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-21103063

ABSTRACT

BACKGROUND: Avian reoviruses replicate in viral factories, which are dense cytoplasmic compartments established by protein-protein interactions. The non-structural protein muNS forms the factory scaffold that attracts other viral components in a controlled fashion. To create such a three-dimensional network, muNS uses several different self-interacting domains. METHODOLOGY/PRINCIPAL FINDINGS: In this study we have devised a strategy to identify muNS regions containing self-interacting domains, based on the capacity of muNS-derived inclusions to recruit muNS fragments. The results revealed that the muNS region consisting of residues 477-542 was recruited with the best efficiency, and this raised the idea of using this fragment as a molecular tag for delivering foreign proteins to muNS inclusions. By combining such tagging system with our previously established method for purifying muNS inclusions from baculovirus-infected insect cells, we have developed a novel protein purification protocol. CONCLUSIONS/SIGNIFICANCE: We show that our tagging and inclusion-targeting system can be a simple, versatile and efficient method for immobilizing and purifying active proteins expressed in baculovirus-infected cells. We also demonstrate that muNS inclusions can simultaneously recruit several tagged proteins, a finding which may be used to generate protein complexes and create multiepitope particulate material for immunization purposes.


Subject(s)
Immobilized Proteins/metabolism , Inclusion Bodies, Viral/metabolism , Orthoreovirus, Avian/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Binding Sites/genetics , Cell Line , Cells, Cultured , Chick Embryo , Fibroblasts/cytology , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immobilized Proteins/genetics , Immunoblotting , Microscopy, Fluorescence , Orthoreovirus, Avian/genetics , Protein Interaction Mapping/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spodoptera , Transfection , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification
14.
J Virol ; 84(9): 4289-301, 2010 May.
Article in English | MEDLINE | ID: mdl-20181708

ABSTRACT

Members of the genus Orthoreovirus replicate in cytoplasmic inclusions termed viral factories. Compelling evidence suggests that the nonstructural protein microNS forms the matrix of the factories and recruits specific viral proteins to these structures. In the first part of this study, we analyzed the properties of avian reovirus factories and microNS-derived inclusions and found that they are nonaggresome cytoplasmic globular structures not associated with the cytoskeleton which do not require an intact microtubule network for formation and maturation. We next investigated the capacity of avian reovirus microNS to form inclusions in transfected and baculovirus-infected cells. Our results showed that microNS is the main component of the inclusions formed by recombinant baculovirus expression. This, and the fact that microNS is able to self-associate inside the cell, suggests that microNS monomers contain all the interacting domains required for inclusion formation. Examination of the inclusion-forming capacities of truncated microNS versions allowed us to identify the region spanning residues 448 to 635 of microNS as the smallest that was inclusion competent, although residues within the region 140 to 380 seem to be involved in inclusion maturation. Finally, we investigated the roles that four different motifs present in microNS(448-635) play in inclusion formation, and the results suggest that the C-terminal tail domain is a key determinant in dictating the initial orientation of monomer-to-monomer contacts to form basal oligomers that control inclusion shape and inclusion-forming efficiency. Our results contribute to an understanding of the generation of structured protein aggregates that escape the cellular mechanisms of protein recycling.


Subject(s)
Inclusion Bodies, Viral/metabolism , Microtubules/metabolism , Orthoreovirus, Avian/physiology , Protein Interaction Domains and Motifs , Protein Multimerization , Viral Nonstructural Proteins/metabolism , Animals , Binding Sites , Cells, Cultured , Chick Embryo , Fibroblasts/virology , Protein Interaction Mapping
15.
J Virol ; 83(19): 10163-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19640987

ABSTRACT

Avian reovirus sigmaA is a double-stranded RNA (dsRNA)-binding protein that has been shown to stabilize viral core particles and to protect the virus against the antiviral action of interferon. To continue with the characterization of this viral protein, we have investigated its intracellular distribution in avian cells. Most sigmaA accumulates into cytoplasmic viral factories of infected cells, and yet a significant fraction was detected in the nucleolus. The protein also localizes in the nucleolus of transfected cells, suggesting that nucleolar targeting is not facilitated by the viral infection or by viral factors. Assays performed in both intact cells and digitonin-permeabilized cells demonstrate that sigmaA is able to enter the nucleus via a nucleoporin-dependent nondiffusional mechanism that does not require added cytosolic factors or energy input. These results indicate that sigmaA by itself is able to penetrate into the nucleus using a process that is mechanistically different from the classical nuclear localization signal/importin pathway. On the other hand, two sigmaA arginines that are necessary for dsRNA binding are also required for nucleolar localization, suggesting that dsRNA-binding and nucleolar targeting are intimately linked properties of the viral protein.


Subject(s)
Cell Nucleolus/metabolism , Orthoreovirus, Avian/metabolism , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Viral Core Proteins/metabolism , Viral Core Proteins/physiology , Animals , Cell Line , Chick Embryo , Cytoplasm/metabolism , Cytosol/metabolism , Digitonin/pharmacology , Karyopherins/metabolism , Microscopy, Fluorescence/methods , Nuclear Localization Signals/metabolism , Recombinant Proteins/chemistry , Subcellular Fractions/metabolism
16.
J Virol ; 82(22): 11208-16, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18799570

ABSTRACT

Avian reovirus, an important avian pathogen, expresses eight structural and four nonstructural proteins. The structural sigmaA protein is a major component of the inner capsid, clamping together lambdaA building blocks. sigmaA has also been implicated in the resistance of avian reovirus to the antiviral action of interferon by strongly binding double-stranded RNA in the host cell cytoplasm and thus inhibiting activation of the double-stranded RNA-dependent protein kinase. We have solved the structure of bacterially expressed sigmaA by molecular replacement and refined it using data to 2.3-A resolution. Twelve sigmaA molecules are present in the P1 unit cell, arranged as two short double helical hexamers. A positively charged patch is apparent on the surface of sigmaA on the inside of this helix and mutation of either of two key arginine residues (Arg155 and Arg273) within this patch abolishes double-stranded RNA binding. The structural data, together with gel shift assay, electron microscopy, and sedimentation velocity centrifugation results, provide evidence for cooperative binding of sigmaA to double-stranded RNA. The minimal length of double-stranded RNA required for sigmaA binding was observed to be 14 to 18 bp.


Subject(s)
Orthoreovirus, Avian/chemistry , RNA-Binding Proteins/chemistry , Viral Core Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Electrophoretic Mobility Shift Assay , Microscopy, Electron , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Orthoreovirus, Avian/ultrastructure , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Alignment , Ultracentrifugation , Viral Core Proteins/ultrastructure
17.
Article in English | MEDLINE | ID: mdl-17565188

ABSTRACT

The avian reovirus protein sigmaA plays a dual role: it is a structural protein forming part of the transcriptionally active core, but it has also been implicated in the resistance of the virus to interferon by strongly binding double-stranded RNA and thus inhibiting the double-stranded RNA-dependent protein kinase. The sigmaA protein has been crystallized from solutions containing ammonium sulfate at pH values around 6. Crystals belonging to space group P1, with unit-cell parameters a = 103.2, b = 129.9, c = 144.0 A, alpha = 93.8, beta = 105.1, gamma = 98.2 degrees were grown and a complete data set has been collected to 2.3 A resolution. The self-rotation function suggests that sigmaA may form symmetric arrangements in the crystals.


Subject(s)
Orthoreovirus, Avian/chemistry , RNA-Binding Proteins/chemistry , Viral Core Proteins/chemistry , Crystallization , Crystallography, X-Ray , Protein Conformation
18.
Virus Res ; 123(2): 105-19, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17018239

ABSTRACT

Avian reoviruses are important pathogens that cause considerable losses to the poultry industry, but they have been poorly characterized at the molecular level in the past, mostly because they have been considered to be very similar to the well-studied mammalian reoviruses. Studies performed over the last 20 years have revealed that avian reoviruses have unique properties and activities, different to those displayed by their mammalian counterparts, and of considerable interest to molecular virologists. Notably, the avian reovirus S1 gene is unique, in that it is a functional tricistronic gene that possesses three out-of-phase and partially overlapping open reading frames; the identification of the mechanisms that govern the initiation of translation of the three S1 cistrons, and the study of the properties and activities displayed by their encoded proteins, are particularly interesting areas of research. For instance, avian reoviruses are one of the few nonenveloped viruses that cause cell-cell fusion, and their fusogenic phenotype has been associated with a nonstructural 10 kDa transmembrane protein, which is expressed by the second cistron of the S1 gene; the small size of this atypical fusion protein offers an interesting model for studying the mechanisms of cell-cell fusion and for identifying fusogenic domains. Finally, avian reoviruses are highly resistant to interferon, and therefore they may be useful for investigating the mechanisms and strategies that viruses utilize to counteract the antiviral actions of interferons.


Subject(s)
Orthoreovirus, Avian/chemistry , Orthoreovirus, Avian/physiology , Animals , Apoptosis , Cell Physiological Phenomena , Cells/virology , Genome, Viral , Molecular Weight , Reoviridae Infections/virology , Viral Proteins/chemistry , Viral Proteins/physiology , Virion/chemistry , Virion/physiology , Virus Replication
19.
J Mol Biol ; 354(1): 137-49, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16236316

ABSTRACT

Avian reovirus fibre, a homo-trimer of the sigmaC protein, is responsible for primary host cell attachment. The protein expressed in bacteria forms elongated fibres comprised of a carboxy-terminal globular head domain and a slender shaft, and partial proteolysis yielded a carboxy-terminal protease-stable domain that was amenable to crystallisation. Here, we show that this fragment retains receptor-binding capability and report its structure, solved using two-wavelength anomalous diffraction and refined using data collected from three different crystal forms at 2.1 angstroms, 2.35 angstroms and 3.0 angstroms resolution. The carboxy-terminal globular domain has a beta-barrel fold with the same overall topology as the mammalian reovirus fibre (sigma1). However, the monomers of the sigmaC trimer show a more splayed-out arrangement than in the sigma1 structure. Also resolved are two triple beta-spiral repeats of the shaft or stalk domain. The presence in the sequence of heptad repeats amino-terminal to these triple beta-spiral repeats suggests that the unresolved portion of the shaft domain contains a triple alpha-helical coiled-coil structure. Implications for the function and stability of the sigmaC protein are discussed.


Subject(s)
Capsid Proteins/chemistry , Orthoreovirus, Avian/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Capsid Proteins/metabolism , Crystallization , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Receptors, Virus/metabolism , Sequence Alignment
20.
J Gen Virol ; 86(Pt 4): 1159-1169, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784910

ABSTRACT

The avian reovirus non-structural protein sigma NS has previously been shown to bind single-stranded (ss) RNA in vitro in a sequence-independent manner. The results of the present study further reveal that sigma NS binds poly(A), poly(U) and ssDNA, but not poly(C), poly(G) or duplex nucleic acids, suggesting that sigma NS has some nucleotide-sequence specificity for ssRNA binding. The current findings also show that sigma NS is present in large ribonucleoprotein complexes in the cytoplasm of avian reovirus-infected cells, indicating that it exists in intimate association with ssRNAs in vivo. Removal of RNA from the complexes generates a sigma NS protein form that sediments between 4.5 and 7 S, suggesting that RNA-free sigma NS associates into small oligomers. Expression and purification of recombinant sigma NS in insect cells allowed us to generate specific antibodies and to perform a variety of assays. The results of these assays revealed that: (i) RNA-free sigma NS exists as homodimers and homotrimers; (ii) the minimum RNA size for sigma NS binding is between 10 and 20 nt; (iii) sigma NS does not have a preference for viral mRNA sequences; and (iv) its RNA-binding activity is conformation-dependent. Baculovirus expression of point and deletion sigma NS mutants in insect cells showed that the five conserved basic amino acids that are important for RNA binding and ribonucleoprotein-complex formation are dispersed throughout the entire sigma NS sequence, suggesting that this protein binds ssRNA through conformational domains. Finally, the properties of the avian reovirus protein sigma NS are compared with those of its mammalian reovirus counterpart.


Subject(s)
Orthoreovirus, Avian/pathogenicity , RNA, Viral/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Chick Embryo , Fibroblasts , Molecular Sequence Data , Mutagenesis , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoproteins/metabolism , Spodoptera , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...