Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Virol ; 87(11): 6211-20, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536662

ABSTRACT

The rotavirus (RV) genome consists of 11 segments of double-stranded RNA (dsRNA). Typically, each segment contains 5' and 3' untranslated regions (UTRs) that flank an open reading frame (ORF) encoding a single protein. RV variants with segments of atypical size owing to sequence rearrangements have been described. In many cases, the rearrangement originates from a partial head-to-tail sequence duplication that initiates after the stop codon of the ORF, leaving the protein product of the segment unaffected. To probe the limits of the RV genome to accommodate additional genetic sequence, we used reverse genetics to insert duplications (analogous to synthetic rearrangements) and heterologous sequences into the 3' UTR of the segment encoding NSP2 (gene 8). The approach allowed the recovery of recombinant RVs that contained sequence duplications (up to 200 bp) and heterologous sequences, including those for FLAG, the hepatitis C virus E2 epitope, and the internal ribosome entry site of cricket paralysis virus. The recombinant RVs grew to high titer (>10(7) PFU/ml) and remained genetically stable during serial passage. Despite their longer 3' UTRs, rearranged RNAs of recombinant RVs expressed wild-type levels of protein in vivo. Competitive growth experiments indicated that, unlike RV segments with naturally occurring sequence duplications, genetically engineered segments were less efficiently packaged into progeny viruses. Thus, features of naturally occurring rearranged segments, other than their increased length, contribute to their enhanced packaging phenotype. Our results define strategies for developing recombinant RVs as expression vectors, potentially leading to next-generation RV vaccines that induce protection against other infectious agents.


Subject(s)
Genome, Viral , Recombination, Genetic , Rotavirus/genetics , 3' Untranslated Regions , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Genetic Engineering , Macaca mulatta , Molecular Sequence Data , RNA, Viral/genetics , Reverse Genetics , Sequence Analysis, DNA
2.
J Gen Virol ; 94(Pt 6): 1296-1300, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23426355

ABSTRACT

Infectious entry of the nonenveloped rotavirus virion requires proteolysis of the spike protein VP4 to mediate conformational changes associated with membrane penetration. We sequenced and characterized an isolate that was cultured in the absence of trypsin and found that it is more resistant to proteolysis than WT virus. A substitution mutation abrogates one of the defined trypsin-cleavage sites, suggesting that blocking proteolysis at this site reduces the overall kinetics of proteolysis. Kinetic analysis of the membrane penetration-associated conformational change indicated that the 'fold-back' of the mutant spike protein is slower than that of WT. Despite these apparent biochemical defects, the mutant virus replicates in an identical manner to the WT virus. These findings enhance an understanding of VP4 functions and establish new strategies to interrogate rotavirus cell entry.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/metabolism , Mutation , Rotavirus Infections/enzymology , Rotavirus Infections/virology , Rotavirus/physiology , Trypsin/metabolism , Amino Acid Sequence , Animals , Host-Pathogen Interactions , Humans , Macaca mulatta , Molecular Sequence Data , Protein Processing, Post-Translational , Rotavirus/chemistry , Rotavirus/genetics , Sequence Alignment , Virus Internalization , Virus Replication
3.
Methods ; 59(2): 199-206, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22687622

ABSTRACT

Effective methods to engineer the segmented, double-stranded RNA genomes of Reoviridae viruses have only recently been developed. Mammalian orthoreoviruses (MRV) and bluetongue virus (BTV) can be recovered from entirely recombinant reagents, significantly improving the capacity to study the replication, pathogenesis, and transmission of these viruses. Conversely, rotaviruses (RVs), which are the major etiological agent of severe gastroenteritis in infants and children, have thus far only been modified using single-segment replacement methods. Reoviridae reverse genetics techniques universally rely on site-specific initiation of transcription by T7 RNA polymerase to generate the authentic 5' end of recombinant RNA segments, but they vary in how the RNAs are introduced into cells: recombinant BTV is recovered by transfection of in vitro transcribed RNAs, whereas recombinant MRV and RV RNAs are transcribed intracellularly from transfected plasmid cDNAs. Additionally, several parameters have been identified in each system that are essential for recombinant virus recovery. Generating recombinant BTV requires the use of 5' capped RNAs and is enhanced by multiple rounds of RNA transfection, suggesting that translation of viral proteins is likely the rate-limiting step. For RV, the efficiency of recovery is almost entirely dependent on the strength of the selection mechanism used to isolate the single-segment recombinant RV from the unmodified helper virus. The reverse genetics methods for BTV and RV are presented and compared to the previously described MRV methods. Analysis and comparison of each method suggest several key lines of research that might lead to a reverse genetics system for RV, analogous to those used for MRV and BTV.


Subject(s)
Reoviridae/genetics , Reverse Genetics/methods , Transcription, Genetic/genetics , Animals , Humans , Reoviridae/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Curr Opin Virol ; 2(4): 373-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22595300

ABSTRACT

Rotaviruses are members of the Reoviridae family of non-enveloped viruses and important etiologic agents of acute gastroenteritis in infants and young children. In recent years, high-resolution structures of triple-layered rotavirus virions and the constituent proteins have provided valuable insights into functions. Of note, structural studies have revealed the position of the viral RNA-dependent RNA polymerase, VP1, within the inner capsid, which in turn provides clues about the location of the viral capping machinery and the route of viral transcript egress. Mechanisms by which the viral spike protein, VP4, mediates receptor binding and membrane penetration have also been aided by high-resolution structural studies. Future work may serve to fill the remaining gaps in understanding of rotavirus particle structure and function.


Subject(s)
Capsid Proteins/metabolism , Gastroenteritis/virology , Rotavirus Infections/virology , Rotavirus/physiology , Animals , Capsid Proteins/genetics , Humans , Protein Binding , Rotavirus/genetics
5.
Nat Rev Microbiol ; 10(3): 165-77, 2012 Jan 23.
Article in English | MEDLINE | ID: mdl-22266782

ABSTRACT

Viral replication is rapid and robust, but it is far from a chaotic process. Instead, successful production of infectious progeny requires that events occur in the correct place and at the correct time. Rotaviruses (segmented double-stranded RNA viruses of the Reoviridae family) seem to govern their replication through ordered disassembly and assembly of a triple-layered icosahedral capsid. In recent years, high-resolution structural data have provided unprecedented insight into these events. In this Review, we explore the current understanding of rotavirus replication and how it compares to replication of other Reoviridae family members.


Subject(s)
Rotavirus/physiology , Virus Assembly , Virus Replication , Capsid/metabolism , Genome, Viral , Reoviridae/physiology
6.
J Virol ; 85(20): 10509-17, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849465

ABSTRACT

Antibodies that neutralize rotavirus infection target outer coat proteins VP4 and VP7 and inhibit viral entry. The structure of a VP7-Fab complex (S. T. Aoki, et al., Science 324:1444-1447, 2009) led us to reclassify epitopes into two binding regions at inter- and intrasubunit boundaries of the calcium-dependent trimer. It further led us to show that antibodies binding at the intersubunit boundary inhibit uncoating of the virion outer layer. We have now tested representative antibodies for each of the defined structural epitope regions and find that antibodies recognizing epitopes in either binding region neutralize by cross-linking VP7 trimers. Antibodies that bind at the intersubunit junction neutralize as monovalent Fabs, while those that bind at the intrasubunit region require divalency. The VP7 structure has also allowed us to design a disulfide cross-linked VP7 mutant which recoats double-layered particles (DLPs) as efficiently as does wild-type VP7 but which yields particles defective in cell entry as determined both by lack of infectivity and by loss of α-sarcin toxicity in the presence of recoated particles. We conclude that dissociation of the VP7 trimer is an essential step in viral penetration into cells.


Subject(s)
Antigens, Viral/metabolism , Capsid Proteins/metabolism , Rotavirus/physiology , Virus Internalization , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Capsid Proteins/chemistry , Capsid Proteins/immunology , Cell Line , Disulfides/metabolism , Macaca mulatta
7.
Proc Natl Acad Sci U S A ; 107(43): 18652-7, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937889

ABSTRACT

Current methods for engineering the segmented double-stranded RNA genome of rotavirus (RV) are limited by inefficient recovery of the recombinant virus. In an effort to expand the utility of RV reverse genetics, we developed a method to recover recombinant viruses in which independent selection strategies are used to engineer single-gene replacements. We coupled a mutant SA11 RV encoding a temperature-sensitive (ts) defect in the NSP2 protein with RNAi-mediated degradation of NSP2 mRNAs to isolate a virus containing a single recombinant gene that evades both selection mechanisms. Recovery is rapid and simple; after two rounds of selective passage the recombinant virus reaches titers of ≥10(4) pfu/mL. We used this reverse genetics method to generate a panel of viruses with chimeric NSP2 genes. For one of the chimeric viruses, the introduced NSP2 sequence was obtained from a pathogenic, noncultivated human RV isolate, demonstrating that this reverse genetics system can be used to study the molecular biology of circulating RVs. Combining characterized RV ts mutants and validated siRNA targets should permit the extension of this "two-hit" reverse genetics methodology to other RV genes. Furthermore, application of a dual selection strategy to previously reported reverse genetics methods for RV may enhance the efficiency of recombinant virus recovery.


Subject(s)
Genetic Engineering/methods , Rotavirus/genetics , Amino Acid Sequence , Base Sequence , Cell Line , DNA, Viral/genetics , Genes, Viral , Humans , Molecular Sequence Data , Mutation , RNA Interference , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombination, Genetic , Selection, Genetic , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Temperature , Viral Nonstructural Proteins/genetics
8.
J Virol ; 84(12): 6200-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20375171

ABSTRACT

Experiments in cell-free systems have demonstrated that the VP5 cleavage fragment of the rotavirus spike protein, VP4, undergoes a foldback rearrangement that translocates three clustered hydrophobic loops from one end of the molecule to the other. This conformational change resembles the foldback rearrangements of enveloped virus fusion proteins. By recoating rotavirus subviral particles with recombinant VP4 and VP7, we tested the effects on cell entry of substituting hydrophilic for hydrophobic residues in the clustered VP5 loops. Several of these mutations decreased the infectivity of recoated particles without preventing either recoating or folding back. In particular, the V391D mutant had a diminished capacity to interact with liposomes when triggered to fold back by serial protease digestion in solution, and particles recoated with this mutant VP4 were 10,000-fold less infectious than particles recoated with wild-type VP4. Particles with V391D mutant VP4 attached normally to cells and internalized efficiently, but they failed in the permeabilization step that allows coentry of the toxin alpha-sarcin. These findings indicate that the hydrophobicity of the VP5 apex is required for membrane disruption during rotavirus cell entry.


Subject(s)
Mutation , Rotavirus/physiology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Internalization , Animals , Cell Line , Hydrophobic and Hydrophilic Interactions , Macaca mulatta , Rotavirus/chemistry , Rotavirus/genetics , Viral Nonstructural Proteins/metabolism
9.
Viruses ; 2(6): 1340-1346, 2010 Jun.
Article in English | MEDLINE | ID: mdl-21994683

ABSTRACT

A recently solved structure of the aquareovirus virion (Zhang, X; Jin, L.; Fang, Q; Hui, W.H.; Zhou Z.H. 3.3 Å Cryo-EM Structure of a Nonenveloped Virus Reveals a Priming Mechanism for Cell Entry. Cell2010, 141, 472-482 [1]) provides new insights into the order of entry events, as well as confirming and refining several aspects of the entry mechanism, for aquareovirus and the related orthoreovirus. In particular, the structure provides evidence of a defined order for the progressive proteolytic cleavages of myristoylated penetration protein VP5 that prime the virion for membrane penetration. These observations reinforce the concept that, much like enveloped viruses, nonenveloped virions often undergo priming events that lead to a meta-stable state, preparing the virus for membrane penetration under the appropriate circumstances. In addition, this and other recent studies highlight the increasing power of electron cryomicroscopy to analyze large, geometrically regular structures, such as icosahedral viruses, at atomic resolution.

10.
J Virol ; 84(4): 1764-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20007281

ABSTRACT

During rotavirus entry, a virion penetrates a host cell membrane, sheds its outer capsid proteins, and releases a transcriptionally active subviral particle into the cytoplasm. VP5, the rotavirus protein believed to interact with the membrane bilayer, is a tryptic cleavage product of the outer capsid spike protein, VP4. When a rotavirus particle uncoats, VP5 folds back, in a rearrangement that resembles the fusogenic conformational changes in enveloped-virus fusion proteins. We present direct experimental evidence that this rearrangement leads to membrane binding. VP5 does not associate with liposomes when mounted as part of the trypsin-primed spikes on intact virions, nor does it do so after it has folded back into a stably trimeric, low-energy state. But it does bind liposomes when they are added to virions before uncoating, and VP5 rearrangement is then triggered by addition of EDTA. The presence of liposomes during the rearrangement enhances the otherwise inefficient VP5 conformational change. A VP5 fragment, VP5CT, produced from monomeric recombinant VP4 by successive treatments with chymotrypsin and trypsin, also binds liposomes only when the proteolysis proceeds in their presence. A monoclonal antibody that neutralizes infectivity by blocking a postattachment entry event also blocks VP5 liposome association. We propose that VP5 binds lipid bilayers in an intermediate conformational state, analogous to the extended intermediate conformation of enveloped-virus fusion proteins.


Subject(s)
Rotavirus/chemistry , Viral Nonstructural Proteins/chemistry , Animals , Antibodies, Neutralizing , Cell Line , Chymotrypsin , Lipid Bilayers/chemistry , Liposomes/chemistry , Macaca mulatta , Models, Biological , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rotavirus/genetics , Rotavirus/physiology , Trypsin , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virus Internalization
11.
J Virol ; 83(21): 11372-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19692464

ABSTRACT

Trypsin primes rotavirus for efficient infectivity by cleaving the spike protein, VP4, into VP8* and VP5*. A recombinant VP5* fragment has a trimeric, folded-back structure. Comparison of this structure with virion spikes suggests that a rearrangement, analogous to those of enveloped virus fusion proteins, may mediate membrane penetration by rotavirus during entry. To detect this inferred rearrangement of virion-associated authentic VP5*, we raised conformation-specific monoclonal antibodies against the recombinant VP5* fragment in its putative post-membrane penetration conformation. Using one of these antibodies, we demonstrate that rotavirus uncoating triggers a conformational change in the cleaved VP4 spike to yield rearranged VP5*.


Subject(s)
Protein Structure, Quaternary , Rotavirus/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Rotavirus/ultrastructure , Viral Nonstructural Proteins/genetics , Virion/metabolism , Virion/ultrastructure , Virus Internalization
12.
Science ; 324(5933): 1444-7, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19520960

ABSTRACT

Rotavirus outer-layer protein VP7 is a principal target of protective antibodies. Removal of free calcium ions (Ca2+) dissociates VP7 trimers into monomers, releasing VP7 from the virion, and initiates penetration-inducing conformational changes in the other outer-layer protein, VP4. We report the crystal structure at 3.4 angstrom resolution of VP7 bound with the Fab fragment of a neutralizing monoclonal antibody. The Fab binds across the outer surface of the intersubunit contact, which contains two Ca2+ sites. Mutations that escape neutralization by other antibodies suggest that the same region bears the epitopes of most neutralizing antibodies. The monovalent Fab is sufficient to neutralize infectivity. We propose that neutralizing antibodies against VP7 act by stabilizing the trimer, thereby inhibiting the uncoating trigger for VP4 rearrangement. A disulfide-linked trimer is a potential subunit immunogen.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Capsid Proteins/chemistry , Capsid Proteins/immunology , Immunoglobulin Fab Fragments/immunology , Rotavirus/chemistry , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Viral/chemistry , Antibodies, Viral/metabolism , Antigens, Viral/genetics , Antigens, Viral/metabolism , Binding Sites , Binding Sites, Antibody , Calcium/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Crystallography, X-Ray , Epitopes/immunology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Neutralization Tests , Protein Folding , Protein Multimerization , Protein Structure, Tertiary , Protein Subunits , Recombinant Proteins/chemistry , Rotavirus/immunology , Serotyping
13.
J Virol ; 80(22): 11293-304, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16971442

ABSTRACT

Assembly of the rotavirus outer capsid is the final step of a complex pathway. In vivo, the later steps include a maturational membrane penetration that is dependent on the scaffolding activity of a viral nonstructural protein. In vitro, simply adding the recombinant outer capsid proteins VP4 and VP7 to authentic double-layered rotavirus subviral particles (DLPs) in the presence of calcium and acidic pH increases infectivity by a factor of up to 10(7), yielding particles as infectious as authentic purified virions. VP4 must be added before VP7 for high-level infectivity. Steep dependence of infectious recoating on VP4 concentration suggests that VP4-VP4 interactions, probably oligomerization, precede VP4 binding to particles. Trypsin sensitivity analysis identifies two populations of VP4 associated with recoated particles: properly mounted VP4 that can be specifically primed by trypsin, and nonspecifically associated VP4 that is degraded by trypsin. A full complement of properly assembled VP4 is not required for efficient infectivity. Minimal dependence of recoating on VP7 concentration suggests that VP7 binds DLPs with high affinity. The parameters for efficient recoating and the characterization of recoated particles suggest a model in which, after a relatively weak interaction between oligomeric VP4 and DLPs, VP7 binds the particles and locks VP4 in place. Recoating will allow the use of infectious modified rotavirus particles to explore rotavirus assembly and cell entry and could lead to practical applications in novel immunization strategies.


Subject(s)
Antigens, Viral/metabolism , Capsid Proteins/metabolism , Capsid/metabolism , Rotavirus/pathogenicity , Virus Assembly , Animals , Antigens, Viral/genetics , Capsid Proteins/genetics , Cell Line , Mice , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rotavirus/genetics , Rotavirus/physiology , Rotavirus/ultrastructure , Virion/metabolism , Virion/ultrastructure , Virus Assembly/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...