Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 114(41): E8731-E8740, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973853

ABSTRACT

Computer design and chemical synthesis generated viable variants of poliovirus type 1 (PV1), whose ORF (6,189 nucleotides) carried up to 1,297 "Max" mutations (excess of overrepresented synonymous codon pairs) or up to 2,104 "SD" mutations (randomly scrambled synonymous codons). "Min" variants (excess of underrepresented synonymous codon pairs) are nonviable except for P2Min, a variant temperature-sensitive at 33 and 39.5 °C. Compared with WT PV1, P2Min displayed a vastly reduced specific infectivity (si) (WT, 1 PFU/118 particles vs. P2Min, 1 PFU/35,000 particles), a phenotype that will be discussed broadly. Si of haploid PV presents cellular infectivity of a single genotype. We performed a comprehensive analysis of sequence and structures of the PV genome to determine if evolutionary conserved cis-acting packaging signal(s) were preserved after recoding. We showed that conserved synonymous sites and/or local secondary structures that might play a role in determining packaging specificity do not survive codon pair recoding. This makes it unlikely that numerous "cryptic, sequence-degenerate, dispersed RNA packaging signals mapping along the entire viral genome" [Patel N, et al. (2017) Nat Microbiol 2:17098] play the critical role in poliovirus packaging specificity. Considering all available evidence, we propose a two-step assembly strategy for +ssRNA viruses: step I, acquisition of packaging specificity, either (a) by specific recognition between capsid protein(s) and replication proteins (poliovirus), or (b) by the high affinity interaction of a single RNA packaging signal (PS) with capsid protein(s) (most +ssRNA viruses so far studied); step II, cocondensation of genome/capsid precursors in which an array of hairpin structures plays a role in virion formation.


Subject(s)
Genome, Viral , Poliomyelitis/virology , Poliovirus/genetics , Poliovirus/pathogenicity , Virion/genetics , Virus Assembly , Virus Replication , A549 Cells , HeLa Cells , Humans , Phenotype , Poliomyelitis/genetics , RNA, Viral
2.
J Virol ; 90(14): 6174-6186, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27076638

ABSTRACT

UNLABELLED: The specificity of encapsidation of C-cluster enteroviruses depends on an interaction between capsid proteins and nonstructural protein 2C(ATPase) In particular, residue N252 of poliovirus 2C(ATPase) interacts with VP3 of coxsackievirus A20, in the context of a chimeric virus. Poliovirus 2C(ATPase) has important roles both in RNA replication and encapsidation. In this study, we searched for additional sites in 2C(ATPase), near N252, that are required for encapsidation. Accordingly, segments adjacent to N252 were analyzed by combining triple and single alanine mutations to identify residues required for function. Two triple alanine mutants exhibited defects in RNA replication. The remaining two mutations, located in secondary structures in a predicted three-dimensional model of 2C(ATPase), caused lethal growth phenotypes. Most single alanine mutants, derived from the lethal variants, were either quasi-infectious and yielded variants with wild-type (wt) or temperature-sensitive (ts) growth phenotypes or had a lethal growth phenotype due to defective RNA replication. The K259A mutation, mapping to an α helix in the predicted structure of 2C(ATPase), resulted in a cold-sensitive virus. In vivo protein synthesis and virus production were strikingly delayed at 33°C relative to the wt, suggesting a defect in uncoating. Studies with a reporter virus indicated that this mutant is also defective in encapsidation at 33°C. Cell imaging confirmed a much-reduced production of K259A mature virus at 33°C relative to the wt. In conclusion, we have for the first time linked a cold-sensitive encapsidation defect in 2C(ATPase) (K259A) to a subsequent delay in uncoating of the virus particle at 33°C during the next cycle of infection. IMPORTANCE: Enterovirus morphogenesis, which involves the encapsidation of newly made virion RNA, is a process still poorly understood. Elucidation of this process is important for future drug development for a large variety of diseases caused by these agents. We have previously shown that the specificity of encapsidation of poliovirus and of C-cluster coxsackieviruses, which are prototypes of enteroviruses, is dependent on an interaction of capsid proteins with the multifunctional nonstructural protein 2C(ATPase) In this study, we have searched for residues in poliovirus 2C(ATPase), near a presumed capsid-interacting site, important for encapsidation. An unusual cold-sensitive mutant of 2C(ATPase) possessed a defect in encapsidation at 37°C and subsequently in uncoating during the next cycle of infection at 33°C. These studies not only reveal a new site in 2C(ATPase) that is involved in encapsidation but also identify a link between encapsidation and uncoating.


Subject(s)
Capsid/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutation/genetics , Poliomyelitis/pathology , Poliovirus/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Uncoating , Amino Acid Sequence , Amino Acid Substitution , HeLa Cells , Host-Pathogen Interactions , Humans , Mutagenesis, Site-Directed , Phenotype , Poliomyelitis/genetics , Poliomyelitis/virology , Poliovirus/enzymology , RNA, Viral/genetics , Sequence Homology, Amino Acid , Virus Assembly , Virus Replication
3.
Virology ; 484: 80-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26074065

ABSTRACT

Enteroviruses (EV) uridylylate a peptide, VPg, as the first step in their replication. VPgpUpU, found free in infected cells, serves as the primer for RNA elongation. The abilities of four polymerases (3D(pol)), from EV-species A-C, to uridylylate VPgs that varied by up to 60% of their residues were compared. Each 3D(pol) was able to uridylylate all five VPgs using polyA RNA as template, while showing specificity for its own genome encoded peptide. All 3D(pol) uridylylated a consensus VPg representing the physical chemical properties of 31 different VPgs. Thus the residues required for uridylylation and the enzymatic mechanism must be similar in diverse EV. As VPg-binding sites differ in co-crystal structures, the reaction is probably done by a second 3D(pol) molecule. The conservation of polymerase residues whose mutation reduces uridylylation but not RNA elongation is compared.


Subject(s)
Enterovirus/physiology , Protein Processing, Post-Translational , Uridine Triphosphate/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Enterovirus/enzymology , Humans , Models, Molecular , Protein Conformation , Substrate Specificity , Viral Nonstructural Proteins/chemistry
4.
Virus Res ; 206: 12-26, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-25592245

ABSTRACT

Plus strand RNA viruses use different mechanisms to initiate the synthesis of their RNA chains. The Picornaviridae family constitutes a large group of plus strand RNA viruses that possess a small terminal protein (VPg) covalently linked to the 5'-end of their genomes. The RNA polymerases of these viruses use VPg as primer for both minus and plus strand RNA synthesis. In the first step of the initiation reaction the RNA polymerase links a UMP to the hydroxyl group of a tyrosine in VPg using as template a cis-replicating element (cre) positioned in different regions of the viral genome. In this review we will summarize what is known about the initiation reaction of protein-primed RNA synthesis by the RNA polymerases of the Picornaviridae. As an example we will use the RNA polymerase of poliovirus, the prototype of Picornaviridae. We will also discuss models of how these nucleotidylylated protein primers might be used, together with viral and cellular replication proteins and other cis-replicating RNA elements, during minus and plus strand RNA synthesis.


Subject(s)
Picornaviridae/physiology , RNA, Viral/metabolism , Transcription Initiation, Genetic , Virus Replication , Models, Biological , Nucleic Acid Conformation , Protein Binding , RNA Folding , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism
5.
Microbiol Mol Biol Rev ; 78(3): 418-37, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25184560

ABSTRACT

The Picornaviridae represent a large family of small plus-strand RNA viruses that cause a bewildering array of important human and animal diseases. Morphogenesis is the least-understood step in the life cycle of these viruses, and this process is difficult to study because encapsidation is tightly coupled to genome translation and RNA replication. Although the basic steps of assembly have been known for some time, very few details are available about the mechanism and factors that regulate this process. Most of the information available has been derived from studies of enteroviruses, in particular poliovirus, where recent evidence has shown that, surprisingly, the specificity of encapsidation is governed by a viral protein-protein interaction that does not involve an RNA packaging signal. In this review, we make an attempt to summarize what is currently known about the following topics: (i) encapsidation intermediates, (ii) the specificity of encapsidation (iii), viral and cellular factors that are required for encapsidation, (iv) inhibitors of encapsidation, and (v) a model of enterovirus encapsidation. Finally, we compare some features of picornavirus morphogenesis with those of other plus-strand RNA viruses.


Subject(s)
Picornaviridae Infections/virology , Picornaviridae/physiology , Virus Assembly , Animals , Antiviral Agents/pharmacology , Capsid/physiology , Capsid/ultrastructure , Genome, Viral , Host-Pathogen Interactions , Humans , Morphogenesis , Picornaviridae/drug effects , Picornaviridae/ultrastructure , RNA, Viral/physiology
6.
PLoS Pathog ; 10(4): e1004052, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24722315

ABSTRACT

Glutathione (GSH) is the most abundant cellular thiol playing an essential role in preserving a reduced cellular environment. Cellular GSH levels can be efficiently reduced by the GSH biosynthesis inhibitor, L-buthionine sulfoximine (BSO). The aim of our study was to determine the role of GSH in the growth of two C-cluster enteroviruses, poliovirus type 1 (PV1) and coxsackievirus A20 (CAV20). Our results show that the growth of both PV1 and CAV20 is strongly inhibited by BSO and can be partially reversed by the addition of GSH. BSO has no effect on viral protein synthesis or RNA replication but it strikingly reduces the accumulation of 14S pentamers in infected cells. GSH-pull down assays show that GSH directly interacts with capsid precursors and mature virus made in the absence of BSO whereas capsid precursors produced under GSH-depletion do not bind to GSH. In particular, the loss of binding of GSH may debilitate the stability of 14S pentamers, resulting in their failure to assemble into mature virus. Immunofluorescence cell imaging demonstrated that GSH-depletion did not affect the localization of viral capsid proteins to the replication complex. PV1 BSO resistant (BSOr) mutants evolved readily during passaging of the virus in the presence of BSO. Structural analyses revealed that the BSOr mutations, mapping to VP1 and VP3 capsid proteins, are primarily located at protomer/protomer interfaces. BSOr mutations might, in place of GSH, aid the stability of 14S particles that is required for virion maturation. Our observation that BSOr mutants are more heat resistant and need less GSH than wt virus to be protected from heat inactivation suggests that they possess a more stable capsid. We propose that the role of GSH during enterovirus morphogenesis is to stabilize capsid structures by direct interaction with capsid proteins both during and after the formation of mature virus particles.


Subject(s)
Capsid/metabolism , Enterovirus C, Human/physiology , Enterovirus Infections/metabolism , Glutathione/metabolism , Virus Assembly/physiology , Glutathione/antagonists & inhibitors , HeLa Cells , Humans
7.
J Gen Virol ; 95(Pt 6): 1255-1265, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24558221

ABSTRACT

The morphogenesis of viruses belonging to the genus Enterovirus in the family Picornaviridae is still poorly understood despite decades-long investigations. However, we recently provided evidence that 2C(ATPase) gives specificity to poliovirus encapsidation through an interaction with capsid protein VP3. The polypeptide 2C(ATPase) is a highly conserved non-structural protein of enteroviruses with important roles in RNA replication, encapsidation and uncoating. We have identified a site (K279/R280) near the C terminus of the polypeptide that is required for morphogenesis. The aim of the current project was to search for additional functional sites near the C terminus of the 2C(ATPase) polypeptide, with particular interest in those that are required for encapsidation. We selected for analysis a cysteine-rich site of the polypeptide and constructed four mutants in which cysteines or a histidine was changed to an alanine. The RNA transcripts were transfected into HeLa cells yielding two lethal, one temperature-sensitive and one quasi-infectious mutants. All four mutants exhibited normal protein translation in vitro and three of them possessed severe RNA replication defects. The quasi-infectious mutant (C286A) yielded variants with a pseudo-reversion at the original site (A286D), but some also contained one additional mutation: A138V or M293V. The temperature-sensitive mutant (C272A/H273A) exhibited an encapsidation and possibly also an uncoating defect at 37 °C. Variants of this mutant revealed suppressor mutations at three different sites in the 2C(ATPase) polypeptide: A138V, M293V and K295R. We concluded that the cysteine-rich site near the C terminus of 2C(ATPase) is involved in encapsidation, possibly through an interaction with an upstream segment located between boxes A and B of the nucleotide-binding domain.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Poliovirus/growth & development , Poliovirus/physiology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Binding Sites/genetics , Carrier Proteins/genetics , Conserved Sequence , Genes, Viral , HeLa Cells , Humans , Molecular Sequence Data , Morphogenesis/genetics , Mutation , Phenotype , Poliovirus/genetics , Protein Biosynthesis , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA, Viral/biosynthesis , RNA, Viral/genetics , Sequence Homology, Amino Acid , Suppression, Genetic , Viral Nonstructural Proteins/genetics , Virus Assembly/genetics
8.
Proc Natl Acad Sci U S A ; 109(36): 14301-7, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22886087

ABSTRACT

Genomes of RNA viruses contain multiple functional RNA elements required for translation or RNA replication. We use unique approaches to identify functional RNA elements in the coding sequence of poliovirus (PV), a plus strand RNA virus. The general method is to recode large segments of the genome using synonymous codons, such that protein sequences, codon use, and codon pair bias are conserved but the nucleic acid sequence is changed. Such recoding does not affect the growth of PV unless it destroys the sequence/structure of a functional RNA element. Using genetic analyses and a method called "signal location search," we detected two unique functionally redundant RNA elements (α and ß), each about 75 nt long and separated by 150 nt, in the 3'-terminal coding sequence of RNA polymerase, 3D(pol). The presence of wild type (WT) α or ß was sufficient for the optimal growth of PV, but the alteration of both segments in the same virus yielded very low titers and tiny plaques. The nucleotide sequences and predicted RNA structures of α and ß have no apparent resemblance to each other. In α, we narrowed down the functional domain to a 48-nt-long, highly conserved segment. The primary determinant of function in ß is a stable and highly conserved hairpin. Reporter constructs showed that the α- and ß-segments are required for RNA replication. Recoding offers a unique and effective method to search for unknown functional RNA elements in coding sequences of RNA viruses, particularly if the signals are redundant in function.


Subject(s)
Computer-Aided Design , DNA-Directed RNA Polymerases/genetics , Genetic Engineering/methods , Poliovirus/genetics , RNA, Viral/genetics , Virus Replication/genetics , Poliovirus/growth & development , Protein Structure, Tertiary/genetics
9.
J Virol ; 86(18): 9964-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761387

ABSTRACT

Polypeptide 2C(ATPase) is one of the most thoroughly studied but least understood proteins in the life cycle of poliovirus. Within the protein, multiple functional domains important for uncoating, host cell membrane alterations, and RNA replication and encapsidation have previously been identified. In this study, charged to alanine-scanning mutagenesis was used to generate conditional-lethal mutations in hitherto-uncharacterized domains of the 2C(ATPase) polypeptide, particularly those involved in morphogenesis. Adjacent or clustered charged amino acids (2 to 4), scattered along the 2C(ATPase) coding sequence, were replaced with alanines. RNA transcripts of mutant poliovirus cDNA clones were transfected into HeLa cells. Subsequently, 10 lethal, 1 severely temperature-sensitive, 2 quasi-infectious, and 3 wild type-like mutants were identified. Using a luciferase-containing reporter virus, we demonstrated RNA replication defects in all lethal and quasi-infectious mutants. Temperature-sensitive mutants were defective in RNA replication only at the restricted temperatures. Furthermore, we characterized a quasi-infectious mutant (K(6)A/K(7)A) that produced a suppressor mutation (G(1)R) and a novel 2B^2C(ATPase) cleavage site (Q^R). Surprisingly, this cleavage site mutation did not interfere with normal processing of the polyprotein. These mutants have led to the identification of several new sites within the 2C(ATPase) polypeptide that are required for RNA replication. In addition, analysis of the suppressor mutants has revealed a new domain near the C terminus of 2C(ATPase) that is involved in encapsidation, possibly achieved through interaction with an amino acid sequence between NTP binding motifs A and B of 2C(ATPase). Most importantly, the identification of suppressor mutations in both 2C(ATPase) and the capsid domains (VP1 and VP3) of poliovirus has confirmed that an interaction between 2C(ATPase) and capsid proteins is involved in viral morphogenesis.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Capsid Proteins/genetics , Capsid Proteins/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Poliovirus/genetics , Poliovirus/physiology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Adenosine Triphosphatases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Capsid Proteins/chemistry , Carrier Proteins/chemistry , Conserved Sequence , HeLa Cells , Humans , Molecular Sequence Data , Morphogenesis , Mutagenesis, Site-Directed , Phenotype , Poliovirus/growth & development , Protein Structure, Tertiary , Viral Nonstructural Proteins/chemistry , Virus Assembly/genetics , Virus Assembly/physiology , Virus Replication/genetics , Virus Replication/physiology
10.
J Virol ; 86(4): 1999-2010, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22156535

ABSTRACT

Polioviruses (PVs) carrying a reporter gene are useful tools for studies of virus replication, particularly if the viral chimeras contain the polyprotein that provides all of the proteins necessary for a complete replication cycle. Replication in HeLa cells of a previously constructed poliovirus expressing the gene for Renilla luciferase (RLuc) fused to the N terminus of the polyprotein H(2)N-RLuc-P1-P2-P3-COOH (P1, structural domain; P2 and P3, nonstructural domains) led to the deletion of RLuc after only one passage. Here we describe a novel poliovirus chimera that expresses Gaussia luciferase (GLuc) inserted into the polyprotein between P1 and P2 (N(2)H-P1-GLuc-P2-P3-COOH). This chimera, termed PV-GLuc, replicated to 10% of wild-type yield. The reporter signal was fully retained for three passages and then gradually lost. After six passages the signal was barely detectable. On further passages, however, the GLuc signal reappeared, and after eight passages it had reached the same levels observed with the original PV-GLuc at the first passage. We demonstrated that this surprising observation was due to coevolution of defective interfering (DI) particles that had lost part or all of the capsid coding sequence (ΔP1-GLuc-P2-P3) and wild-type-like viruses that had lost the GLuc sequence (P1-P2-P3). When used at low passage, PV-GLuc is an excellent tool for studying aspects of genome replication and morphogenesis. The GLuc protein was secreted from mammalian cells but, in agreement with published data, was not secreted from PV-GLuc-infected cells due to poliovirus-induced inhibition of cellular protein secretion. Published evidence indicates that individual expression of enterovirus polypeptide 3A, 2B, or 2BC in COS-1 cells strongly inhibits host protein secretion. In HeLa cells, however, expression of none of the poliovirus polypeptides, either singly or in pairs, inhibited GLuc secretion. Thus, inhibition of GLuc secretion in PV-infected HeLa cells is likely a result of the interaction between several viral and cellular proteins that are different from those in COS-1 cells.


Subject(s)
Biological Evolution , Crustacea/enzymology , Defective Viruses/genetics , Gene Expression , Luciferases/genetics , Poliovirus/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Crustacea/genetics , Defective Viruses/metabolism , Genes, Reporter , Luciferases/metabolism , Poliovirus/metabolism , Polyproteins/genetics , Polyproteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
Annu Rev Microbiol ; 65: 583-609, 2011.
Article in English | MEDLINE | ID: mdl-21756105

ABSTRACT

Owing to known genome sequences, modern strategies of DNA synthesis have made it possible to recreate in principle all known viruses independent of natural templates. We describe the first synthesis of a virus (poliovirus) in 2002 that was accomplished outside living cells. We comment on the reaction of laypeople and scientists to the work, which shaped the response to de novo syntheses of other viruses. We discuss those viruses that have been synthesized since 2002, among them viruses whose precise genome sequence had to be established by painstakingly stitching together pieces of sequence information, and viruses involved in zoonosis. Synthesizing viral genomes provides a powerful tool for studying gene function and the pathogenic potential of these organisms. It also allows modification of viral genomes to an extent hitherto unthinkable. Recoding of poliovirus and influenza virus to develop new vaccine candidates and refactoring the phage T7 DNA genome are discussed as examples.


Subject(s)
Bacteriophage T7/chemistry , DNA, Viral/chemical synthesis , Orthomyxoviridae/chemistry , Poliovirus/chemistry , RNA, Viral/chemical synthesis , Bacteriophage T7/genetics , Bacteriophage T7/physiology , DNA, Viral/genetics , Genes, Synthetic , Genome, Viral , Humans , Orthomyxoviridae/genetics , Orthomyxoviridae/physiology , Poliovirus/genetics , Poliovirus/physiology , RNA, Viral/genetics , Virus Replication
12.
PLoS Pathog ; 6(8): e1001066, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20865167

ABSTRACT

In spite of decades-long studies, the mechanism of morphogenesis of plus-stranded RNA viruses belonging to the genus Enterovirus of Picornaviridae, including poliovirus (PV), is not understood. Numerous attempts to identify an RNA encapsidation signal have failed. Genetic studies, however, have implicated a role of the non-structural protein 2C(ATPase) in the formation of poliovirus particles. Here we report a novel mechanism in which protein-protein interaction is sufficient to explain the specificity in PV encapsidation. Making use of a novel "reporter virus", we show that a quasi-infectious chimera consisting of the capsid precursor of C-cluster coxsackie virus 20 (C-CAV20) and the nonstructural proteins of the closely related PV translated and replicated its genome with wild type kinetics, whereas encapsidation was blocked. On blind passages, encapsidation of the chimera was rescued by a single mutation either in capsid protein VP3 of CAV20 or in 2C(ATPase) of PV. Whereas each of the single-mutation variants expressed severe proliferation phenotypes, engineering both mutations into the chimera yielded a virus encapsidating with wild type kinetics. Biochemical analyses provided strong evidence for a direct interaction between 2C(ATPase) and VP3 of PV and CAV20. Chimeras of other C-CAVs (CAV20/CAV21 or CAV18/CAV20) were blocked in encapsidation (no virus after blind passages) but could be rescued if the capsid and 2C(ATPase) coding regions originated from the same virus. Our novel mechanism explains the specificity of encapsidation without apparent involvement of an RNA signal by considering that (i) genome replication is known to be stringently linked to translation, (ii) morphogenesis is known to be stringently linked to genome replication, (iii) newly synthesized 2C(ATPase) is an essential component of the replication complex, and (iv) 2C(ATPase) has specific affinity to capsid protein(s). These conditions lead to morphogenesis at the site where newly synthesized genomes emerge from the replication complex.


Subject(s)
Capsid Proteins/metabolism , Carrier Proteins/metabolism , Enterovirus/physiology , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Capsid Proteins/genetics , Carrier Proteins/genetics , HeLa Cells , Humans , Immunoprecipitation , Poliovirus/genetics , Poliovirus/metabolism , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics
13.
Biochim Biophys Acta ; 1789(9-10): 495-517, 2009.
Article in English | MEDLINE | ID: mdl-19781674

ABSTRACT

The RNA genomes of plus-strand RNA viruses have the ability to form secondary and higher-order structures that contribute to their stability and to their participation in inter- and intramolecular interactions. Those structures that are functionally important are called cis-acting RNA elements because their functions cannot be complemented in trans. They can be involved not only in RNA/RNA interactions but also in binding of viral and cellular proteins during the complex processes of translation, RNA replication and encapsidation. Most viral cis-acting RNA elements are located in the highly structured 5'- and 3'-nontranslated regions of the genomes but sometimes they also extend into the adjacent coding sequences. In addition, some cis-acting RNA elements are embedded within the coding sequences far away from the genomic ends. Although the functional importance of many of these structures has been confirmed by genetic and biochemical analyses, their precise roles are not yet fully understood. In this review we have summarized what is known about cis-acting RNA elements in nine families of human and animal plus-strand RNA viruses with an emphasis on the most thoroughly characterized virus families, the Picornaviridae and Flaviviridae.


Subject(s)
RNA Viruses/genetics , RNA, Viral/genetics , RNA/chemistry , 3' Untranslated Regions , 5' Untranslated Regions , Animals , Base Sequence , Enhancer Elements, Genetic , Flavivirus/metabolism , Gene Expression Regulation, Viral , Genome, Viral , Humans , Models, Genetic , Molecular Sequence Data , Nucleic Acid Conformation , Picornaviridae/metabolism
14.
J Virol ; 81(20): 11256-66, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17686844

ABSTRACT

Lethal mutagenesis is the mechanism of action of ribavirin against poliovirus (PV) and numerous other RNA viruses. However, there is still considerable debate regarding the mechanism of action of ribavirin against a variety of RNA viruses. Here we show by using T7 RNA polymerase-mediated production of PV genomic RNA, PV polymerase-catalyzed primer extension, and cell-free PV synthesis that a pyrimidine ribonucleoside triphosphate analogue (rPTP) with ambiguous base-pairing capacity is an efficient mutagen of the PV genome. The in vitro incorporation properties of rPTP are superior to ribavirin triphosphate. We observed a log-linear relationship between virus titer reduction and the number of rPMP molecules incorporated. A PV genome encoding a high-fidelity polymerase was more sensitive to rPMP incorporation, consistent with diminished mutational robustness of high-fidelity PV. The nucleoside (rP) did not exhibit antiviral activity in cell culture, owing to the inability of rP to be converted to rPMP by cellular nucleotide kinases. rP was also a poor substrate for herpes simplex virus thymidine kinase. The block to nucleoside phosphorylation could be bypassed by treatment with the P nucleobase, which exhibited both antiviral activity and mutagenesis, presumably a reflection of rP nucleotide formation by a nucleotide salvage pathway. These studies provide additional support for lethal mutagenesis as an antiviral strategy, suggest that rPMP prodrugs may be highly efficacious antiviral agents, and provide a new tool to determine the sensitivity of RNA virus genomes to mutagenesis as well as interrogation of the impact of mutational load on the population dynamics of these viruses.


Subject(s)
Antiviral Agents , Mutagenesis/drug effects , Poliovirus/genetics , Pyrimidines/pharmacology , RNA, Viral/biosynthesis , Genome, Viral , Mutagens/pharmacology , Pyrimidine Nucleotides/metabolism , Pyrimidine Nucleotides/pharmacology , Pyrimidine Nucleotides/therapeutic use , Pyrimidines/metabolism , Pyrimidines/therapeutic use
15.
J Gen Virol ; 88(Pt 8): 2259-2267, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17622630

ABSTRACT

All of the non-structural proteins of poliovirus, including their processing precursors, are involved in the replication of the viral RNA genome. These proteins assemble into a replication complex, which also contains the viral RNA and cellular factors. An understanding of how these viral proteins interact with each other would enhance our understanding of the molecular events occurring during poliovirus infection of the cell. Previously, we have employed the yeast two-hybrid system to construct two separate linkage maps for the polioviral P2 and P3 proteins, respectively. In the present study, we have searched for interacting pairs between the P2 and P3 proteins in a similar inducible yeast two-hybrid system. Although, the primary functions of the proteolytic products of the P2 and P3 domains of the polyprotein in the viral life cycle are different, we observed significant interactions between 2C(ATPase) and 3AB; 2A(pro) and 3A, 3C(pro) or 3D(pol); 2B and 3A or 3AB. All of the interactions were measured in the yeast two-hybrid system by exchanging the interacting pairs on the transcription-activation and DNA-binding constructs. In vitro GST pull-down assay suggested that the 2C(ATPase)/3AB interaction involves both ionic and hydrophobic contacts between the two proteins. The possible biological implication of the interactions observed in the yeast two-hybrid system will be discussed.


Subject(s)
Calcium-Transporting ATPases/metabolism , Poliomyelitis/virology , Poliovirus/genetics , Viral Nonstructural Proteins/metabolism , 3C Viral Proteases , Cysteine Endopeptidases/metabolism , Glutathione Transferase/metabolism , Membrane Proteins/metabolism , Protein Binding , Two-Hybrid System Techniques , Viral Proteins/metabolism , Yeasts
16.
J Virol ; 81(18): 10017-28, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17609276

ABSTRACT

The 5' nontranslated region of poliovirus RNA contains two highly structured regions, the cloverleaf (CL) and the internal ribosomal entry site (IRES). A cellular protein, the poly(rC) binding protein (PCBP), has been reported to interact with the CL either alone or in combination with viral protein 3CD(pro). The formation of the ternary complex is essential for RNA replication and, hence, viral proliferation. PCBP also interacts with stem-loop IV of the IRES, an event critical for the initiation of cap-independent translation. Until recently, no special function was assigned to a spacer region (nucleotides [nt] 89 to 123) located between the CL and the IRES. However, on the basis of our discovery that this region strongly affects the neurovirulent phenotype of poliovirus, we have embarked upon genetic and biochemical analyses of the spacer region, focusing on two clusters of C residues (C(93-95) and C(98-100)) that are highly conserved among entero- and rhinoviruses. Replacement of all six C residues with A residues had no effect on translation in vitro but abolished RNA replication, leading to a lethal growth phenotype of the virus in HeLa cells. Mutation of the first group of C residues (C(93-95)) resulted in slower viral growth, whereas the C(98-100)A change had no significant effect on viability. Genetic analyses of the C-rich region by extensive mutagenesis and analyses of revertants revealed that two consecutive C residues (C(94-95)) were sufficient to promote normal growth of the virus. However, there was a distinct position effect of the preferred C residues. A 142-nt-long 5'-terminal RNA fragment including the CL and spacer sequences efficiently bound PCBP, whereas no PCBP binding was observed with the CL (nt 1 to 88) alone. Binding of PCBP to the 142-nt fragment was completely ablated after the two C clusters in the spacer were mutated to A clusters. In contrast, the same mutations had no effect on the binding of 3CD(pro) to the 142-nt RNA fragment. Stepwise replacement of the C residues with A residues resulted in impaired replication that covaried with weaker binding of PCBP in vitro. We conclude that PCBP has little, if any, binding affinity for the CL itself (nt 1 to 88) but requires additional nucleotides downstream of the CL for its function as an essential cofactor in poliovirus RNA replication. These data reveal a new essential function of the spacer between the CL and the IRES in poliovirus proliferation.


Subject(s)
5' Untranslated Regions/metabolism , Poliovirus/metabolism , Poly C/metabolism , RNA, Viral/biosynthesis , RNA-Binding Proteins/metabolism , Virus Replication/physiology , 5' Untranslated Regions/genetics , Binding Sites/genetics , HeLa Cells , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nucleic Acid Conformation , Poliovirus/genetics , Poly C/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Rhinovirus/genetics , Rhinovirus/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
17.
Biochemistry ; 46(17): 5185-99, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17417822

ABSTRACT

Replication of poliovirus RNA takes place on the cytoplasmic surface of membranous vesicles that form after infection of the host cell. It is generally accepted that RNA polymerase 3D(pol) interacts with membranes in a complex with viral protein 3AB, which binds to membranes by means of a hydrophobic anchor sequence that is located near the C-terminus of the 3A domain. In this study, we used fluorescence and fluorescence quenching methods to define the topography of the anchor sequence in the context of 3A and 3AB proteins inserted in model membranes. Mutants with a single tryptophan near the center of the anchor sequence but lacking Trp elsewhere in 3A/3AB were constructed which, after the emergence of suppressor mutations, replicated well in HeLa cells. When a peptide containing the mutant anchor sequence was incorporated in model membrane vesicles, measurements of Trp depth within the lipid bilayer indicated formation of a transmembrane topography. However, rather than the 22-residue length predicted from hydrophobicity considerations, the transmembrane segment had an effective length of 16 residues, such that Gln64 likely formed the N-terminal boundary. Analogous experiments using full-length proteins bound to preformed model membrane vesicles showed that the anchor sequence formed a mixture of transmembrane and nontransmembrane topographies in the 3A protein but adopted only the nontransmembrane configuration in the context of 3AB protein. Studies of the function of 3A/3AB inserted into model membrane vesicles showed that membrane-bound 3AB is highly efficient in stimulating the activity of 3D(pol) in vitro while membrane-bound 3A totally lacks this activity. Moreover, in vitro uridylylation reactions showed that membrane-bound 3AB is not a substrate for 3D(pol), but free VPg released by cleavage of 3AB with proteinase 3CD(pro) could be uridylylated.


Subject(s)
Poliovirus/metabolism , RNA, Viral/biosynthesis , Viral Proteins/metabolism , Acrylamide/chemistry , Amino Acid Sequence , Base Sequence , Binding Sites , DNA Primers , Molecular Sequence Data , Mutagenesis , Poliovirus/genetics , Protein Binding , Spectrometry, Fluorescence , Transcription, Genetic , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/isolation & purification
18.
J Virol ; 81(11): 5669-84, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17360746

ABSTRACT

Poliovirus (PV) VPg is a genome-linked protein that is essential for the initiation of viral RNA replication. It has been well established that RNA replication is initiated when a molecule of UMP is covalently linked to the hydroxyl group of a tyrosine (Y3) in VPg by the viral RNA polymerase 3D(pol), but it is not yet known whether the substrate for uridylylation in vivo is the free peptide itself or one of its precursors. The aim of this study was to use complementation analyses to obtain information about the true in vivo substrate for uridylylation by 3D(pol). Previously, it was shown that a VPg mutant, in which tyrosine 3 and threonine 4 were replaced by phenylalanine and alanine (3F4A), respectively, was nonviable. We have now tested whether wild-type forms of proteins 3B, 3BC, 3BCD, 3AB, 3ABC, and P3 provided either in trans or in cis could rescue the replication defect of the VPg(3F4A) mutations in the PV polyprotein. Our results showed that proteins 3B, 3BC, 3BCD, and P3 were unable to complement the RNA replication defect in dicistronic PV or dicistronic luciferase replicons in vivo. However, cotranslation of the P3 precursor protein allowed rescue of RNA replication of the VPg(3F4A) mutant in an in vitro cell-free translation-RNA replication system, but only poor complementation was observed when 3BC, 3AB, 3BCD, or 3ABC proteins were cotranslated in the same assay. Interestingly, only protein 3AB but not 3B and 3BC, when provided in cis by insertion of a wild-type 3AB coding sequence between the P2 and P3 domains of the polyprotein, supported the replication of the mutated genome in vivo. Elimination of cleavage between 3A and 3B in the complementing 3AB protein, however, led to a complete lack of RNA replication. Our results suggest that (i) VPg has to be delivered to the replication complex in the form of a large protein precursor (P3) to be fully functional in replication; (ii) the replication complex formed during PV replication in vivo is essentially inaccessible to proteins provided in trans, even if the complementing protein is translated from a different cistron of the same RNA genome; (iii) 3AB is the most likely precursor of VPg; and (iv) Y3 of VPg has an essential function in RNA replication in the context of both VPg and 3AB.


Subject(s)
Peptide Fragments/physiology , Poliovirus/physiology , Protein Precursors/physiology , RNA, Viral/biosynthesis , Tyrosine/physiology , Viral Proteins/physiology , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Poliovirus/chemistry , Protein Precursors/chemistry , Tyrosine/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/genetics
19.
J Virol ; 80(22): 11343-54, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16971430

ABSTRACT

Replication of the plus-stranded RNA genome of hepatitis C virus (HCV) occurs in a membrane-bound replication complex consisting of viral and cellular proteins and viral RNA. NS5B, the RNA polymerase of HCV, is anchored to the membranes via a C-terminal 20-amino-acid-long hydrophobic domain, which is flanked on each side by a highly conserved positively charged arginine. Using a genotype 1b subgenomic replicon (V. Lohmann, F. Korner, J. O. Koch, U. Herian, L. Theilmann, and R. Bartensclager, Science 285:110-113, 1999), we determined the effect of mutations of some highly conserved residues in this domain. The replacement of arginine 570 with alanine completely abolished the colony-forming ability by the replicon, while a R591A change was found to be highly detrimental to replication, viability, and membrane binding by the mutant NS5B protein. Mutations of two other highly conserved amino acids (L588A and P589A) reduced but did not eliminate colony formation. It was of interest, if specific amino acid residues play a role in membrane anchoring of NS5B and replication, to determine whether a complete exchange of the NS5B hydrophobic domain with a domain totally unrelated to NS5B would ablate replication. We selected the 22-amino-acid-long hydrophobic domain of poliovirus polypeptide 3A that is known to adopt a transmembrane configuration, thereby anchoring 3A to membranes. Surprisingly, either partial or full replacement of the NS5B hydrophobic domain with the anchor sequences of poliovirus polypeptide 3A resulted in the replication of replicons whose colony-forming abilities were reduced compared to that of the wild-type replicon. Upon continued passage of the replicon in Huh-7 cells in the presence of neomycin, the replication efficiency of the replicon increased. However, the sequence of the poliovirus polypeptide 3A hydrophobic domain, in the context of the subgenomic HCV replicon, was stably maintained throughout 40 passages. Our results suggest that anchoring NS5B to membranes is necessary but that the amino acid sequence of the anchor per se does not require HCV origin. This suggests that specific interactions between the NS5B hydrophobic domain and other membrane-bound factors may not play a decisive role in HCV replication.


Subject(s)
Hepacivirus/physiology , Viral Core Proteins/genetics , Viral Core Proteins/physiology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Amino Acid Substitution , Cell Line, Tumor , Cell Membrane/chemistry , Cell Nucleus/chemistry , Cytoplasm/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Microbial Viability , Microscopy, Fluorescence , Models, Molecular , Mutagenesis, Site-Directed , Mutation, Missense , Poliovirus , Protein Structure, Tertiary , Recombinant Proteins , Viral Nonstructural Proteins/chemistry , Viral Plaque Assay , Virus Replication
20.
Virol J ; 2: 86, 2005 Nov 21.
Article in English | MEDLINE | ID: mdl-16300678

ABSTRACT

Poliovirus protein 3CDpro possesses both proteinase and RNA binding activities, which are located in the 3Cpro domain of the protein. The RNA polymerase (3Dpol) domain of 3CDpro modulates these activities of the protein. We have recently shown that the level of 3CDpro in HeLa cell-free in vitro translation-RNA replication reactions is suboptimal for efficient virus production. However, the addition of either 3CDpro mRNA or of purified 3CDpro protein to in vitro reactions, programmed with viral RNA, results in a 100-fold increase in virus yield. Mutational analyses of 3CDpro indicated that RNA binding by the 3Cpro domain and the integrity of interface I in the 3Dpol domain of the protein are both required for function. The aim of these studies was to determine the exact step or steps at which 3CDpro enhances virus yield and to determine the mechanism by which this occurs. Our results suggest that the addition of extra 3CDpro to in vitro translation RNA-replication reactions results in a mild enhancement of both minus and plus strand RNA synthesis. By examining the viral particles formed in the in vitro reactions on sucrose gradients we determined that 3CDpro has only a slight stimulating effect on the synthesis of capsid precursors but it strikingly enhances the maturation of virus particles. Both the stimulation of RNA synthesis and the maturation of the virus particles are dependent on the presence of an intact RNA binding site within the 3Cpro domain of 3CDpro. In addition, the integrity of interface I in the 3Dpol domain of 3CDpro is required for efficient production of mature virus. Surprisingly, plus strand RNA synthesis and virus production in in vitro reactions, programmed with full-length transcript RNA, are not enhanced by the addition of extra 3CDpro. Our results indicate that the stimulation of RNA synthesis and virus maturation by 3CDpro in vitro is dependent on the presence of a VPg-linked RNA template.


Subject(s)
Cysteine Endopeptidases/metabolism , Poliovirus/metabolism , RNA, Viral/biosynthesis , Viral Proteins/metabolism , Virus Assembly , 3C Viral Proteases , Cell-Free System , Gene Expression Regulation, Viral , HeLa Cells , Humans , Protein Biosynthesis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...