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










Publication year range
1.
J Mol Graph Model ; 78: 61-73, 2017 11.
Article in English | MEDLINE | ID: mdl-29032052

ABSTRACT

Crystal structure of the protrusion domain (P-domain) of the grouper nervous necrosis virus (GNNV) shows the presence of three-fold trimeric protrusions with two asymmetrical calcium cations along the non-crystallographic three-fold axis. The trimeric interaction natures of the interacting residues and the calcium cations with the neighboring residues within the trimeric interface have been studied by the quantum theory of atoms in molecules (QTAIM) and natural bond orbital (NBO) analyses in the framework of the density-functional theory (DFT) approach. The results revealed that residues Leu259, Val274, Trp280, and Gln322 of subunit A, Arg261, Asp275, Ala277, and Gln322 of subunit B, Leu259, Asp260, Arg261, Ala277, Val278, and Leu324 of subunit C are the main residues involved in the trimeric interactions. Charge-dipole, dipole-dipole, and hydrogen bonding interactions make the significant contributions to these trimeric interactions. Among different interacting residues within trimeric interface, residue pair Arg261 B-Leu259C forms the strongest hydrogen bond inside the interface between subunits B and C. It was also found that calcium cations interact with residues Asp273, Val274, and Asp275 of subunits A, B, and C through charge-charge and charge transfer interactions.


Subject(s)
Calcium/chemistry , Molecular Conformation , Orthoreovirus/chemistry , Viral Proteins/chemistry , Amino Acids/chemistry , Cations , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Orthoreovirus/genetics , Quantum Theory
2.
Oncotarget ; 7(48): 79814-79827, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27806335

ABSTRACT

Reoviruses are potential anticancer agents due to their ability to induce cell death in tumor cells. Grass carp reovirus (GCRV) is one of the best characterized models on reovirus pathogenesis in vitro. However, there is little known about how SUMOylation affects reovirus pathogenesis. The SUMO conjugating enzyme 9 (Ubc9) determines the targets of SUMOylation. Here, the protein interactions between reovirus outer fiber proteins, specifically GCRV-104 VP55, and Ubc9 were probed using a yeast two-hybrid system. The N-terminal coiled-coil domain of VP55, containing a single lysine residue, was responsible for the interaction between VP55 and Ubc9 in yeast. In solid phase binding assays, a single amino acid mutation (K87R) prevented Ubc9 from binding to VP55. Overexpression of Ubc9 enhanced GCRV-104 infection efficiency, and knockdown of Ubc9 in CIK cells inhibited viral replication, which suggested that Ubc9 was a proviral factor. Furthermore, Ubc9 was shown to bind outer fiber proteins from type II GCRV, avian reovirus and mammalian reovirus in yeast. To our knowledge, this is the first study to show that Ubc9 binds to reovirus outer-fiber proteins and likely contributes to efficient orthoreovirus replication. These results suggest that SUMOylation modifications could be targeted to improve the therapeutic efficacy of oncolytic reovirus.


Subject(s)
Orthoreovirus , Sumoylation , Ubiquitin-Conjugating Enzymes/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Carps/virology , Fish Diseases/metabolism , Fish Diseases/virology , Hemorrhage/metabolism , Hemorrhage/virology , Orthoreovirus/chemistry , Orthoreovirus/metabolism , Phylogeny , Protein Binding , Protein Processing, Post-Translational , Reoviridae Infections/metabolism , Reoviridae Infections/pathology , Reoviridae Infections/veterinary , Sequence Alignment
3.
Biochim Biophys Acta ; 1848(2): 408-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450808

ABSTRACT

The reovirus p10 fusion-associated small transmembrane (FAST) proteins are the smallest known membrane fusion proteins, and evolved specifically to mediate cell-cell, rather than virus-cell, membrane fusion. The 36-40-residue ectodomains of avian reovirus (ARV) and Nelson Bay reovirus (NBV) p10 contain an essential intramolecular disulfide bond required for both cell-cell fusion and lipid mixing between liposomes. To more clearly define the functional, biochemical and biophysical features of this novel fusion peptide, synthetic peptides representing the p10 ectodomains of ARV and NBV were analyzed by solution-state NMR spectroscopy, circular dichroism spectroscopy, fluorescence spectroscopy-based hydrophobicity analysis, and liposome binding and fusion assays. Results indicate that disulfide bond formation promotes exposure of hydrophobic residues, as indicated by bis-ANS binding and time-dependent peptide aggregation under aqueous conditions, implying the disulfide bond creates a small, geometrically constrained, cystine noose. Noose formation is required for peptide partitioning into liposome membranes and liposome lipid mixing, and electron microscopy revealed that liposome-liposome fusion occurs in the absence of liposome tubulation. In addition, p10 fusion peptide activity, but not membrane partitioning, is dependent on membrane cholesterol.


Subject(s)
Cholesterol/chemistry , Cystine/chemistry , Liposomes/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Hydrophobic and Hydrophilic Interactions , Membrane Fusion , Molecular Sequence Data , Orthoreovirus/chemistry , Orthoreovirus, Avian/chemistry , Protein Structure, Secondary , Viral Fusion Proteins/chemical synthesis
4.
J Virol ; 85(15): 7483-95, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21593159

ABSTRACT

Baboon reovirus (BRV) is a member of the fusogenic subgroup of orthoreoviruses. Unlike most other members of its genus, BRV lacks S-segment coding sequences for the outer fiber protein that binds to cell surface receptors. It shares this lack with aquareoviruses, which constitute a related genus and are also fusogenic. We used electron cryomicroscopy and three-dimensional image reconstruction to determine the BRV virion structure at 9.0-Å resolution. The results show that BRV lacks a protruding fiber at its icosahedral 5-fold axes or elsewhere. The results also show that BRV is like nonfusogenic mammalian and fusogenic avian orthoreoviruses in having 150 copies of the core clamp protein, not 120 as in aquareoviruses. On the other hand, there are no hub-and-spoke complexes attributable to the outer shell protein in the P2 and P3 solvent channels of BRV, which makes BRV like fusogenic avian orthoreoviruses and aquareoviruses but unlike nonfusogenic mammalian orthoreoviruses. The outermost "flap" domains of the BRV core turret protein appear capable of conformational variability within the virion, a trait previously unseen among other ortho- and aquareoviruses. New cDNA sequence determinations for the BRV L1 and M2 genome segments, encoding the core turret and outer shell proteins, were helpful for interpreting the structural features of those proteins. Based on these findings, we conclude that the evolution of ortho- and aquareoviruses has included a series of discrete gains or losses of particular components, several of which cross taxonomic boundaries. Gain or loss of adhesion fibers is one of several common themes in double-stranded RNA virus evolution.


Subject(s)
Orthoreovirus/chemistry , Virion/chemistry , Amino Acid Sequence , Animals , Chlorocebus aethiops , Cryoelectron Microscopy , Microscopy, Electron, Transmission , Molecular Sequence Data , Orthoreovirus/physiology , Phylogeny , Protein Conformation , Sequence Homology, Amino Acid , Vero Cells , Virion/genetics , Virion/ultrastructure
5.
J Mol Biol ; 382(1): 213-22, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18625243

ABSTRACT

Grass carp reovirus (GCRV) is a member of the Aquareovirus genus of the family Reoviridae, a large family of double-stranded RNA (dsRNA) viruses infecting plants, insects, fishes and mammals. We report the first subnanometer-resolution three-dimensional structures of both GCRV core and virion by cryoelectron microscopy. These structures have allowed the delineation of interactions among the over 1000 molecules in this enormous macromolecular machine and a detailed comparison with other dsRNA viruses at the secondary-structure level. The GCRV core structure shows that the inner proteins have strong structural similarities with those of orthoreoviruses even at the level of secondary-structure elements, indicating that the structures involved in viral dsRNA interaction and transcription are highly conserved. In contrast, the level of similarity in structures decreases in the proteins situated in the outer layers of the virion. The proteins involved in host recognition and attachment exhibit the least similarities to other members of Reoviridae. Furthermore, in GCRV, the RNA-translocating turrets are in an open state and lack a counterpart for the sigma1 protein situated on top of the close turrets observed in mammalian orthoreovirus. Interestingly, the distribution and the organization of GCRV core proteins resemble those of the cytoplasmic polyhedrosis virus, a cypovirus and the structurally simplest member of the Reoviridae family. Our results suggest that GCRV occupies a unique structure niche between the simpler cypoviruses and the considerably more complex mammalian orthoreovirus, thus providing an important model for understanding the structural and functional conservation and diversity of this enormous family of dsRNA viruses.


Subject(s)
Carps/virology , Reoviridae/ultrastructure , Virion/ultrastructure , Animals , Capsid/ultrastructure , Cell Line , Cryoelectron Microscopy , Dimerization , Models, Molecular , Orthoreovirus/chemistry , Protein Binding , Protein Structure, Quaternary , Reoviridae/chemistry , Viral Proteins/chemistry , Viral Proteins/ultrastructure
6.
J Biol Chem ; 282(24): 17930-40, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17452315

ABSTRACT

Junctional adhesion molecule-A (JAM-A) serves as a serotype-independent receptor for mammalian orthoreoviruses (reoviruses). The membrane-distal immunoglobulin-like D1 domain of JAM-A is required for homodimerization and binding to reovirus attachment protein sigma1. We employed a structure-guided mutational analysis of the JAM-A dimer interface to identify determinants of reovirus binding. We purified mutant JAM-A ectodomains for solution-phase and surface plasmon resonance binding studies and expressed mutant forms of full-length JAM-A in Chinese hamster ovary cells to assess reovirus binding and infectivity. Mutation of residues in the JAM-A dimer interface that participate in salt-bridge or hydrogen-bond interactions with apposing JAM-A monomers abolishes the capacity of JAM-A to form dimers. JAM-A mutants incapable of dimer formation form complexes with the sigma1 head that are indistinguishable from wild-type JAM-A-sigma1 head complexes, indicating that sigma1 binds to JAM-A monomers. Residues Glu(61) and Lys(63) of beta-strand C and Leu(72) of beta-strand C' in the dimer interface are required for efficient JAM-A engagement of strain type 3 Dearing sigma1. Mutation of neighboring residues alters the kinetics of the sigma1-JAM-A binding interaction. Prototype reovirus strains type 1 Lang and type 2 Jones share similar, although not identical, binding requirements with type 3 Dearing. These results indicate that reovirus engages JAM-A monomers via residues found mainly on beta-strands C and C' of the dimer interface and raise the possibility that the distinct disease phenotypes produced in mice following infection with different strains of reovirus are in part attributable to differences in contacts with JAM-A.


Subject(s)
Cell Adhesion Molecules/chemistry , Immunoglobulins/chemistry , Orthoreovirus/metabolism , Protein Structure, Tertiary , Animals , Binding Sites , CHO Cells , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cricetinae , Cricetulus , DNA Mutational Analysis , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Mice , Models, Molecular , Orthoreovirus/chemistry , Point Mutation , Receptors, Cell Surface , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
7.
Mol Cell Proteomics ; 6(6): 1027-38, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17339631

ABSTRACT

Protein-protein associations are vital to cellular functions. Here we describe a helpful new method to demonstrate protein-protein associations inside cells based on the capacity of orthoreovirus protein muNS to form large cytoplasmic inclusions, easily visualized by light microscopy, and to recruit other proteins to these structures in a specific manner. We introduce this technology by the identification of a sixth orthoreovirus protein, RNA-dependent RNA polymerase lambda3, that was recruited to the structures through an association with muNS. We then established the broader utility of this technology by using a truncated, fluorescently tagged form of muNS as a fusion platform to present the mammalian tumor suppressor p53, which strongly recruited its known interactor simian virus 40 large T antigen to the muNS-derived structures. In both examples, we further localized a region of the recruited protein that is key to its recruitment. Using either endogenous p53 or a second fluorescently tagged fusion of p53 with the rotavirus NSP5 protein, we demonstrated p53 oligomerization as well as p53 association with another of its cellular interaction partners, the CREB-binding proteins, within the inclusions. Furthermore using the p53-fused fluorescent muNS platform in conjunction with three-color microscopy, we identified a ternary complex comprising p53, simian virus 40 large T antigen, and retinoblastoma protein. The new method is technically simple, uses commonly available resources, and is adaptable to high throughput formats.


Subject(s)
Orthoreovirus/chemistry , Viral Proteins/metabolism , Animals , Antigens, Polyomavirus Transforming/metabolism , COS Cells , Chlorocebus aethiops , Cytoplasmic Structures/metabolism , Green Fluorescent Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Transport , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
8.
Virology ; 361(2): 412-25, 2007 May 10.
Article in English | MEDLINE | ID: mdl-17208266

ABSTRACT

The 76-kDa mu1 protein of nonfusogenic mammalian reovirus is a major component of the virion outer capsid, which contains 200 mu1 trimers arranged in an incomplete T=13 lattice. In virions, mu1 is largely covered by a second major outer-capsid protein, sigma3, which limits mu1 conformational mobility. In infectious subvirion particles, from which sigma3 has been removed, mu1 is broadly exposed on the surface and can be promoted to rearrange into a protease-sensitive and hydrophobic conformer, leading to membrane perforation or penetration. In this study, mutants that resisted loss of infectivity upon heat inactivation (heat-resistant mutants) were selected from infectious subvirion particles of reovirus strains Type 1 Lang and Type 3 Dearing. All of the mutants were found to have mutations in mu1, and the heat-resistance phenotype was mapped to mu1 by both recoating and reassortant genetics. Heat-resistant mutants were also resistant to rearrangement to the protease-sensitive conformer of mu1, suggesting that heat inactivation is associated with mu1 rearrangement, consistent with published results. Rate constants of heat inactivation were determined, and the dependence of inactivation rate on temperature was consistent with the Arrhenius relationship. The Gibbs free energy of activation was calculated with reference to transition-state theory and was found to be correlated with the degree of heat resistance in each of the analyzed mutants. The mutations are located in upper portions of the mu1 trimer, near intersubunit contacts either within or between trimers in the viral outer capsid. We propose that the mutants stabilize the outer capsid by interfering with unwinding of the mu1 trimer.


Subject(s)
Capsid Proteins/genetics , Hot Temperature , Orthoreovirus/chemistry , Orthoreovirus/physiology , Reoviridae Infections/virology , Animals , Capsid Proteins/chemistry , Cell Line , Models, Molecular , Molecular Weight , Mutation , Orthoreovirus/pathogenicity , Virulence , Virus Inactivation
9.
J Struct Biol ; 157(1): 211-25, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16979906

ABSTRACT

Model-based, three-dimensional (3D) image reconstruction procedures require a starting model to initiate data analysis. We have designed an ab initio method, which we call the random model (RM) method, that automatically generates models to initiate structural analysis of icosahedral viruses imaged by cryo-electron microscopy. The robustness of the RM procedure was demonstrated on experimental sets of images for five representative viruses. The RM method also provides a straightforward way to generate unbiased starting models to derive independent 3D reconstructions and obtain a more reliable assessment of resolution. The fundamental scheme embodied in the RM method should be relatively easy to integrate into other icosahedral software packages.


Subject(s)
Computer Simulation , Imaging, Three-Dimensional/methods , Viruses/chemistry , Algorithms , Animals , Bass/virology , Dengue Virus/chemistry , Electronic Data Processing , Models, Molecular , Nodaviridae/chemistry , Orthoreovirus/chemistry , Paramecium/virology , Phycodnaviridae/chemistry
10.
Structure ; 13(10): 1545-57, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16216585

ABSTRACT

Reovirus is a useful model for addressing the molecular basis of membrane penetration by one of the larger nonenveloped animal viruses. We now report the structure of the reovirus virion at approximately 7.0 A resolution as obtained by electron cryomicroscopy and three-dimensional image reconstruction. Several features of the myristoylated outer capsid protein mu1, not seen in a previous X-ray crystal structure of the mu1-sigma3 heterohexamer, are evident in the virion. These features appear to be important for stabilizing the outer capsid, regulating the conformational changes in mu1 that accompany perforation of target membranes, and contributing directly to membrane penetration during cell entry.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Orthoreovirus/chemistry , Orthoreovirus/ultrastructure , Virion/ultrastructure , Algorithms , Amino Acid Sequence , Animals , Capsid Proteins/isolation & purification , Crystallography, X-Ray , Fourier Analysis , L Cells , Mice , Models, Chemical , Models, Molecular , Molecular Sequence Data , Myristic Acid/metabolism , Orthoreovirus/genetics , Protein Conformation , Protein Structure, Secondary , Sequence Homology, Amino Acid , Virion/growth & development
11.
Virus Res ; 101(1): 15-28, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15010214

ABSTRACT

Orthoreoviruses and Aquareoviruses constitute two respective genera in the family Reoviridae of double-stranded RNA viruses. Orthoreoviruses infect mammals, birds, and reptiles and have a genome comprising 10 RNA segments. Aquareoviruses infect fish and have a genome comprising 11 RNA segments. Despite these differences, recent structural and nucleotide sequence evidence indicate that the proteins of Orthoreoviruses and Aquareoviruses share many similarities. The focus of this review is on the structure and function of the Orthoreovirus core proteins lambda1, lambda2, lambda3, and sigma2, for which X-ray crystal structures have been recently reported. The homologous core proteins in Aquareoviruses are VP3, VP1, VP2, and VP6, respectively. By mapping the locations of conserved residues onto the Orthoreovirus crystal structures, we have found that enzymatic surfaces involved in mRNA synthesis are well conserved between these two groups of viruses, whereas several surfaces involved in protein-protein interactions are not well conserved. Other evidence indicates that the Orthoreovirus mu2 and Aquareovirus VP5 proteins are homologous, suggesting that VP5 is a core protein as mu2 is known to be. These findings provide further evidence that Orthoreoviruses and Aquareoviruses have diverged from a common ancestor and contribute to a growing understanding of the functions of the core proteins in viral mRNA synthesis.


Subject(s)
Orthoreovirus/chemistry , Orthoreovirus/genetics , Reoviridae/chemistry , Reoviridae/genetics , Viral Core Proteins/chemistry , Viral Core Proteins/genetics , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Evolution, Molecular , Models, Molecular , Molecular Sequence Data , Orthoreovirus/metabolism , Protein Conformation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Reoviridae/metabolism , Sequence Homology, Amino Acid , Viral Core Proteins/metabolism
12.
Virology ; 296(1): 24-38, 2002 Apr 25.
Article in English | MEDLINE | ID: mdl-12036315

ABSTRACT

The 144-kDa lambda2 protein, a component of the transcriptionally active reovirus core particle, catalyzes the last three enzymatic activities for formation of the 5' cap 1 structure on the viral plus-strand transcripts. Limited evidence suggests it may also play a role in transcription per se. Particle-associated lambda2 forms pentameric turrets ("spikes") around the fivefold axes of the icosahedral core. To address the requirements for lambda2 in core functions other than the known functions in RNA capping, particles depleted of lambda2 were generated from cores in vitro by a series of treatments involving heat, protease, and ionic detergent. The resulting particles contained less than 5% of pretreatment levels of lambda2 but showed negligible loss of the other four core proteins or the 10 double-stranded RNA genome segments. Transmission cryo-electron microscopy (cryo-TEM) and scanning cryo-electron microscopy demonstrated loss of the lambda2 spikes from these otherwise intact particles. In functional analyses, the "spikeless cores" showed greatly reduced activities not only for RNA capping but also for transcription and nucleoside triphosphate hydrolysis, suggesting enzymatic or structural roles for lambda2 in all these activities. Comparison of the core and spikeless core structures obtained by cryo-TEM and three-dimensional image reconstruction revealed changes in the lambda1 core shell that accompany lambda2 loss, most notably the elimination of small pores that span the shell near the icosahedral fivefold axes. Changes in the shell may explain the reductions in transcriptase-related activities by spikeless cores.


Subject(s)
Orthoreovirus/genetics , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , Viral Core Proteins/metabolism , Cryoelectron Microscopy , Detergents , Endopeptidases , Hot Temperature , Immunoblotting , Models, Molecular , Orthoreovirus/chemistry , Transcription, Genetic , Viral Core Proteins/analysis , Virus Assembly
13.
J Virol ; 74(18): 8472-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954547

ABSTRACT

The reovirus attachment protein, sigma1, is responsible for strain-specific patterns of viral tropism in the murine central nervous system and receptor binding on cultured cells. The sigma1 protein consists of a fibrous tail domain proximal to the virion surface and a virion-distal globular head domain. To better understand mechanisms of reovirus attachment to cells, we conducted studies to identify the region of sigma1 that binds cell surface carbohydrate. Chimeric and truncated sigma1 proteins derived from prototype reovirus strains type 1 Lang (T1L) and type 3 Dearing (T3D) were expressed in insect cells by using a baculovirus vector. Assessment of expressed protein susceptibility to proteolytic cleavage, binding to anti-sigma1 antibodies, and oligomerization indicates that the chimeric and truncated sigma1 proteins are properly folded. To assess carbohydrate binding, recombinant sigma1 proteins were tested for the capacity to agglutinate mammalian erythrocytes and to bind sialic acid presented on glycophorin, the cell surface molecule bound by type 3 reovirus on human erythrocytes. Using a panel of two wild-type and ten chimeric and truncated sigma1 proteins, the sialic acid-binding domain of type 3 sigma1 was mapped to a region of sequence proposed to form the more amino terminal of two predicted beta-sheet structures in the tail. This unit corresponds to morphologic region T(iii) observed in computer-processed electron micrographs of sigma1 protein purified from virions. In contrast, the homologous region of T1L sigma1 sequence was not implicated in carbohydrate binding; rather, sequences in the distal portion of the tail known as the neck were required. Results of these studies demonstrate that a functional receptor-binding domain, which uses sialic acid as its ligand, is contained within morphologic region T(iii) of the type 3 sigma1 tail. Furthermore, our findings indicate that T1L and T3D sigma1 proteins contain different arrangements of receptor-binding domains.


Subject(s)
Capsid Proteins , Carbohydrate Metabolism , Mammalian orthoreovirus 3/metabolism , Orthoreovirus/metabolism , Receptors, Virus/metabolism , Viral Proteins/metabolism , Animals , Baculoviridae/genetics , Cell Line , Glycophorins/chemistry , Hemagglutination , Humans , Insecta/cytology , Mammalian orthoreovirus 3/chemistry , N-Acetylneuraminic Acid/chemistry , Orthoreovirus/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Viral Proteins/chemistry
14.
Nat Struct Biol ; 6(6): 565-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10360362

ABSTRACT

Cytoplasmic polyhedrosis virus (CPV) is unique among the double-stranded RNA viruses of the family Reoviridae in having a single capsid layer. Analysis by cryo-electron microscopy allows comparison of the single shelled CPV and orthoreovirus with the high resolution crystal structure of the inner shell of the bluetongue virus (BTV) core. This suggests that the novel arrangement identified in BTV, of 120 protein subunits in a so-called 'T=2' organization, is a characteristic of the Reoviridae and allows us to delineate structural similarities and differences between two subgroups of the family--the turreted and the smooth-core viruses. This in turn suggests a coherent picture of the structural organization of many dsRNA viruses.


Subject(s)
Orthoreovirus/ultrastructure , RNA Viruses/ultrastructure , RNA, Double-Stranded , Reoviridae/ultrastructure , Bluetongue virus/chemistry , Bluetongue virus/ultrastructure , Capsid/chemistry , Capsid/ultrastructure , Cryoelectron Microscopy , Crystallization , Genome, Viral , Models, Molecular , Orthoreovirus/chemistry , Protein Conformation , RNA Viruses/chemistry , RNA, Double-Stranded/genetics , RNA, Double-Stranded/ultrastructure , RNA, Viral/genetics , RNA, Viral/ultrastructure , Reoviridae/chemistry , Reoviridae/genetics , Viral Core Proteins/chemistry , Viral Core Proteins/ultrastructure
15.
Virology ; 245(1): 33-46, 1998 May 25.
Article in English | MEDLINE | ID: mdl-9614865

ABSTRACT

The structure of mammalian orthoreovirus top component particles, which are profoundly deficient in the content of double-stranded RNA genome, was determined at 30 A resolution by transmission cryoelectron microscopy and three-dimensional image reconstruction. Previously undetected, ordered densities, appearing primarily as pentameric flowers in the reconstruction, were seen to extend 65 A inwardly from the inner capsid at the icosahedral fivefold axes. Identically positioned but lower density elements were observed in two types of partially uncoated top component particles obtained by limited proteolysis. The levels of three inner-capsid proteins-lamda 1, lamda 3, and mu 2-were reduced in concert with the internal densities during proteolytic uncoating. Since lamda 3 contains the catalytic regions of the viral RNA polymerase and since both lamda 1 and mu 2 appear to play roles in transcription or mRNA capping, the internal structures are concluded to be complexes of the viral transcriptase-related enzymes. The findings have implications for the mechanisms of transcription and mRNA capping by orthoreovirus particles.


Subject(s)
Capsid/ultrastructure , Models, Molecular , Orthoreovirus/ultrastructure , Virion/ultrastructure , Animals , Capsid/chemistry , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/ultrastructure , Orthoreovirus/chemistry , Virion/chemistry
20.
J Virol Methods ; 65(2): 159-67, 1997 May.
Article in English | MEDLINE | ID: mdl-9186939

ABSTRACT

A nested polymerase chain reaction (PCR) with subsequent nucleotide sequence analysis identified and differentiated avian reoviruses (ARVs). PCR products amplified from the S1 gene segment of ARV of USA isolates were 738 and 342 bp, respectively. PCR products were conformed by Southern and dot blot hybridizations. The amplified cDNA fragments were cloned into the pUC18 vector and subjected to DNA sequencing. The nucleotide and deduced amino acid sequences of four USA (S1133, 1733, 2408, and CO8) and two Australian isolates (RAM-1 and SOM-4) were compared. Results of paired difference analysis and a predicted dendrogram revealed that USA isolates were closely related, but different from, Australian isolates. The deduced amino acid sequences of the N-terminal region of ARV sigma C showed a heptapeptide repeat of hydrophobic residues in all ARV isolates.


Subject(s)
Orthoreovirus/chemistry , Orthoreovirus/genetics , Polymerase Chain Reaction/methods , Sequence Analysis, DNA , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Chick Embryo , Cloning, Molecular , Fibroblasts , Immunoblotting , Molecular Sequence Data , Orthoreovirus/isolation & purification , Phylogeny , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...