Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nature ; 625(7995): 611-617, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123676

ABSTRACT

The respiratory syncytial virus (RSV) polymerase is a multifunctional RNA-dependent RNA polymerase composed of the large (L) protein and the phosphoprotein (P). It transcribes the RNA genome into ten viral mRNAs and replicates full-length viral genomic and antigenomic RNAs1. The RSV polymerase initiates RNA synthesis by binding to the conserved 3'-terminal RNA promoters of the genome or antigenome2. However, the lack of a structure of the RSV polymerase bound to the RNA promoter has impeded the mechanistic understanding of RSV RNA synthesis. Here we report cryogenic electron microscopy structures of the RSV polymerase bound to its genomic and antigenomic viral RNA promoters, representing two of the first structures of an RNA-dependent RNA polymerase in complex with its RNA promoters in non-segmented negative-sense RNA viruses. The overall structures of the promoter-bound RSV polymerases are similar to that of the unbound (apo) polymerase. Our structures illustrate the interactions between the RSV polymerase and the RNA promoters and provide the structural basis for the initiation of RNA synthesis at positions 1 and 3 of the RSV promoters. These structures offer a deeper understanding of the pre-initiation state of the RSV polymerase and could aid in antiviral research against RSV.


Subject(s)
Promoter Regions, Genetic , RNA-Dependent RNA Polymerase , Respiratory Syncytial Virus, Human , Promoter Regions, Genetic/genetics , Respiratory Syncytial Virus, Human/enzymology , Respiratory Syncytial Virus, Human/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , Virus Replication/genetics , Cryoelectron Microscopy , Subgenomic RNA/biosynthesis , Subgenomic RNA/genetics , Subgenomic RNA/metabolism
2.
Viral Immunol ; 34(1): 18-26, 2021.
Article in English | MEDLINE | ID: mdl-32429800

ABSTRACT

The resolution revolution of cryo-electron microscopy (cryo-EM) has made a significant impact on the structural analysis of the Pneumoviridae multifunctional RNA polymerases. In recent months, several high-resolution structures of apo RNA polymerases of Pneumoviridae, which includes the human respiratory syncytial virus (HRSV) and human metapneumovirus (HMPV), have been determined by single-particle cryo-EM. These structures illustrated high similarities and minor differences between the Pneumoviridae polymerases and revealed the potential mechanisms of the Pneumoviridae RNA synthesis.


Subject(s)
Cryoelectron Microscopy , DNA-Directed RNA Polymerases/ultrastructure , Pneumovirus/enzymology , Humans , Respiratory Syncytial Virus, Human/enzymology
3.
Nat Commun ; 11(1): 368, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953395

ABSTRACT

The respiratory syncytial virus (RSV) RNA polymerase, constituted of a 250 kDa large (L) protein and tetrameric phosphoprotein (P), catalyzes three distinct enzymatic activities - nucleotide polymerization, cap addition, and cap methylation. How RSV L and P coordinate these activities is poorly understood. Here, we present a 3.67 Å cryo-EM structure of the RSV polymerase (L:P) complex. The structure reveals that the RNA dependent RNA polymerase (RdRp) and capping (Cap) domains of L interact with the oligomerization domain (POD) and C-terminal domain (PCTD) of a tetramer of P. The density of the methyltransferase (MT) domain of L and the N-terminal domain of P (PNTD) is missing. Further analysis and comparison with other RNA polymerases at different stages suggest the structure we obtained is likely to be at an elongation-compatible stage. Together, these data provide enriched insights into the interrelationship, the inhibitors, and the evolutionary implications of the RSV polymerase.


Subject(s)
Cryoelectron Microscopy , DNA-Directed RNA Polymerases/chemistry , RNA-Dependent RNA Polymerase/chemistry , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Models, Molecular , Phosphoproteins/chemistry , Protein Conformation , Protein Domains , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Viral Structures
4.
Cell ; 179(1): 193-204.e14, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31495574

ABSTRACT

Numerous interventions are in clinical development for respiratory syncytial virus (RSV) infection, including small molecules that target viral transcription and replication. These processes are catalyzed by a complex comprising the RNA-dependent RNA polymerase (L) and the tetrameric phosphoprotein (P). RSV P recruits multiple proteins to the polymerase complex and, with the exception of its oligomerization domain, is thought to be intrinsically disordered. Despite their critical roles in RSV transcription and replication, structures of L and P have remained elusive. Here, we describe the 3.2-Å cryo-EM structure of RSV L bound to tetrameric P. The structure reveals a striking tentacular arrangement of P, with each of the four monomers adopting a distinct conformation. The structure also rationalizes inhibitor escape mutants and mutations observed in live-attenuated vaccine candidates. These results provide a framework for determining the molecular underpinnings of RSV replication and transcription and should facilitate the design of effective RSV inhibitors.


Subject(s)
Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/ultrastructure , Acetates/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Catalytic Domain , Cryoelectron Microscopy , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Quinolines/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Vaccines/chemistry , Sf9 Cells , Spodoptera , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/drug effects
5.
Viruses ; 11(4)2019 04 04.
Article in English | MEDLINE | ID: mdl-30987343

ABSTRACT

Remdesivir (GS-5734) is a 1'-cyano-substituted adenosine nucleotide analogue prodrug that shows broad-spectrum antiviral activity against several RNA viruses. This compound is currently under clinical development for the treatment of Ebola virus disease (EVD). While antiviral effects have been demonstrated in cell culture and in non-human primates, the mechanism of action of Ebola virus (EBOV) inhibition for remdesivir remains to be fully elucidated. The EBOV RNA-dependent RNA polymerase (RdRp) complex was recently expressed and purified, enabling biochemical studies with the relevant triphosphate (TP) form of remdesivir and its presumptive target. In this study, we confirmed that remdesivir-TP is able to compete for incorporation with adenosine triphosphate (ATP). Enzyme kinetics revealed that EBOV RdRp and respiratory syncytial virus (RSV) RdRp incorporate ATP and remdesivir-TP with similar efficiencies. The selectivity of ATP against remdesivir-TP is ~4 for EBOV RdRp and ~3 for RSV RdRp. In contrast, purified human mitochondrial RNA polymerase (h-mtRNAP) effectively discriminates against remdesivir-TP with a selectivity value of ~500-fold. For EBOV RdRp, the incorporated inhibitor at position i does not affect the ensuing nucleotide incorporation event at position i+1. For RSV RdRp, we measured a ~6-fold inhibition at position i+1 although RNA synthesis was not terminated. Chain termination was in both cases delayed and was seen predominantly at position i+5. This pattern is specific to remdesivir-TP and its 1'-cyano modification. Compounds with modifications at the 2'-position show different patterns of inhibition. While 2'-C-methyl-ATP is not incorporated, ara-ATP acts as a non-obligate chain terminator and prevents nucleotide incorporation at position i+1. Taken together, our biochemical data indicate that the major contribution to EBOV RNA synthesis inhibition by remdesivir can be ascribed to delayed chain termination. The long distance of five residues between the incorporated nucleotide analogue and its inhibitory effect warrant further investigation.


Subject(s)
Adenosine/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/pharmacology , Ebolavirus/enzymology , Enzyme Inhibitors/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Ribonucleotides/pharmacology , Viral Proteins/antagonists & inhibitors , Adenosine/chemistry , Adenosine/metabolism , Adenosine/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Alanine/chemistry , Alanine/metabolism , Alanine/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , DNA-Directed RNA Polymerases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Kinetics , Molecular Structure , Prodrugs , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/enzymology , Respiratory Syncytial Virus, Human/physiology , Ribonucleotides/chemistry , Ribonucleotides/metabolism , Substrate Specificity , Viral Proteins/metabolism
6.
J Med Chem ; 60(6): 2305-2325, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28245119

ABSTRACT

Respiratory syncytial virus (RSV) represents a threat to infants, the elderly, and the immunocompromised. RSV entry blockers are in clinical trials, but escape mutations challenge their potential. In search of RSV inhibitors, we have integrated a signature resistance mutation into a recombinant RSV virus and applied the strain to high-throughput screening. Counterscreening of candidates returned 14 confirmed hits with activities in the nano- to low-micromolar range. All blocked RSV polymerase activity in minigenome assays. Compound 1a (GRP-74915) was selected for development based on activity (EC50 = 0.21 µM, selectivity index (SI) 40) and scaffold. Resynthesis confirmed the potency of the compound, which suppressed viral RNA synthesis in infected cells. However, metabolic testing revealed a short half-life in the presence of mouse hepatocyte fractions. Metabolite tracking and chemical elaboration combined with 3D-quantitative structure-activity relationship modeling yielded analogues (i.e., 8n: EC50 = 0.06 µM, SI 500) that establish a platform for the development of a therapeutic candidate.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Drug Design , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/enzymology , Animals , Antiviral Agents/metabolism , Cell Line , DNA-Directed RNA Polymerases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Mice , Quantitative Structure-Activity Relationship , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
7.
Antiviral Res ; 134: 63-76, 2016 10.
Article in English | MEDLINE | ID: mdl-27575793

ABSTRACT

Worldwide, respiratory syncytial virus (RSV) causes severe disease in infants, the elderly, and immunocompromised people. No vaccine or effective antiviral treatment is available. RSV is a member of the non-segmented, negative-strand (NNS) group of RNA viruses and relies on its RNA-dependent RNA polymerase to transcribe and replicate its genome. Because of its essential nature and unique properties, the RSV polymerase has proven to be a good target for antiviral drugs, with one compound, ALS-8176, having already achieved clinical proof-of-concept efficacy in a human challenge study. In this article, we first provide an overview of the role of the RSV polymerase in viral mRNA transcription and genome replication. We then review past and current approaches to inhibiting the RSV polymerase, including use of nucleoside analogs and non-nucleoside inhibitors. Finally, we consider polymerase inhibitors that hold promise for treating infections with other NNS RNA viruses, including measles and Ebola.


Subject(s)
Antiviral Agents/therapeutic use , DNA-Directed RNA Polymerases/antagonists & inhibitors , Mononegavirales/drug effects , Respiratory Syncytial Virus, Human/drug effects , Antiviral Agents/pharmacology , Clinical Trials as Topic , DNA-Directed RNA Polymerases/drug effects , DNA-Directed RNA Polymerases/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Humans , Measles/drug therapy , Mononegavirales/enzymology , Mononegavirales/genetics , Nucleosides/agonists , RNA, Messenger , RNA-Dependent RNA Polymerase/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/enzymology , Respiratory Syncytial Virus, Human/genetics , Transcription, Genetic , Virus Replication/drug effects
8.
Antiviral Res ; 131: 19-25, 2016 07.
Article in English | MEDLINE | ID: mdl-27059228

ABSTRACT

Respiratory syncytial virus (RSV) infections affect millions of children and adults every year. Despite the significant disease burden, there are currently no safe and effective vaccines or therapeutics. We employed a replicon-based high throughput screen combined with live-virus triaging assays to identify three novel diversity-oriented synthesis-derived scaffolds with activity against RSV. One of these small molecules is shown to target the RSV polymerase (L protein) to inhibit viral replication and transcription; the mechanisms of action of the other small molecules are currently unknown. The compounds described herein may provide attractive inhibitors for lead optimization campaigns.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/methods , High-Throughput Screening Assays/methods , Microbial Sensitivity Tests , Replicon/drug effects , Respiratory Syncytial Virus, Human/drug effects , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Hep G2 Cells , Humans , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/enzymology , Respiratory Syncytial Virus, Human/physiology , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects
9.
J Virol ; 90(10): 4876-4888, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26656699

ABSTRACT

UNLABELLED: Human respiratory syncytial virus (HRSV) is a major cause of serious respiratory tract infection. Treatment options include administration of ribavirin, a purine analog, although the mechanism of its anti-HRSV activity is unknown. We used transcriptome sequencing (RNA-seq) to investigate the genome mutation frequency and viral mRNA accumulation in HRSV-infected cells that were left untreated or treated with ribavirin. In the absence of ribavirin, HRSV-specific transcripts accounted for up to one-third of total RNA reads from the infected-cell RNA population. Ribavirin treatment resulted in a >90% reduction in abundance of viral mRNA reads, while at the same time no such reduction was detected for the abundance of cellular transcripts. The presented data reveal that ribavirin significantly increases the frequency of HRSV-specific RNA mutations, suggesting a direct influence on the fidelity of the HRSV polymerase. The presented data show that transitions and transversions occur during HRSV replication and that these changes occur in hot spots along the HRSV genome. Examination of nucleotide substitution rates in the viral genome indicated an increase in the frequency of transition but not transversion mutations in the presence of ribavirin. In addition, our data indicate that in the continuous cell types used and at the time points analyzed, the abundances of some HRSV mRNAs do not reflect the order in which the mRNAs are transcribed. IMPORTANCE: Human respiratory syncytial virus (HRSV) is a major pediatric pathogen. Ribavirin can be used in children who are extremely ill to reduce the amount of virus and to lower the burden of disease. Ribavirin is used as an experimental therapy with other viruses. The mechanism of action of ribavirin against HRSV is not well understood, although it is thought to increase the mutation rate of the viral polymerase during replication. To investigate this hypothesis, we used a high-resolution approach that allowed us to determine the genetic sequence of the virus to a great depth of coverage. We found that ribavirin did not cause a detectable change in the relative amounts of viral mRNA transcripts. However, we found that ribavirin treatment did indeed cause an increase in the number of mutations, which was associated with a decrease in virus production.


Subject(s)
Antiviral Agents/pharmacology , Mutation , RNA, Viral/genetics , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/physiology , Ribavirin/pharmacology , Transcriptome , Genome, Viral/drug effects , High-Throughput Nucleotide Sequencing/methods , Humans , Interferon-beta/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/enzymology , Respiratory Syncytial Virus, Human/genetics , Transcriptome/drug effects , Transcriptome/genetics , Viral Plaque Assay , Virus Attachment/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects
10.
Proteins ; 72(3): 946-58, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18300250

ABSTRACT

The oligomeric state and the hydrodynamic properties of human respiratory syncytial virus (HRSV) phosphoprotein (P), a known cofactor of the viral RNA-dependent RNA polymerase (L), and a trypsin-resistant fragment (X) that includes its oligomerization domain were analyzed by sedimentation equilibrium and velocity using analytical ultracentrifugation. The results obtained demonstrate that both P and fragment X are homotetrameric with elongated shapes, consistent with electron micrographs of the purified P protein in which thin rod-like molecules of approximately 12.5 +/- 1.0 nm in length were observed. A new chymotrypsin resistant fragment (Y*) included in fragment X has been identified and purified by gel filtration chromatography. Fragment Y* may represent a minimal version of the P oligomerization domain. Thermal denaturation curves based on circular dichroism data of P protein showed a complex behavior. In contrast, melting data generated for fragments X and particularly fragment Y* showed more homogeneous transitions indicative of simpler structures. A three-dimensional model of X and Y* fragments was built based on the atomic structure of the P oligomerization domain of the related Sendai virus, which is in good agreement with the experimental data. This model will be an useful tool to make rational mutations and test the role of specific amino acids in the oligomerization and functional properties of the HRSV P protein.


Subject(s)
Coenzymes/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus, Human/enzymology , Sequence Homology, Amino Acid , Viral Structural Proteins/chemistry , Amino Acid Sequence , Chymotrypsin , Circular Dichroism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Protein Denaturation , Protein Structure, Quaternary , Protein Structure, Tertiary , Temperature , Thermodynamics , Trypsin , Viral Structural Proteins/ultrastructure
11.
Virology ; 330(1): 147-57, 2004 Dec 05.
Article in English | MEDLINE | ID: mdl-15527841

ABSTRACT

The interaction between the respiratory syncytial virus (RSV) polymerase complex and lipid rafts was examined in HEp2 cells. Lipid-raft membranes were prepared from virus-infected cells and their protein content was analysed by Western blotting and mass spectrometry. This analysis revealed the presence of the N, P, L, M2-1 and M proteins. However, these proteins appeared to differ from one another in their association with these structures, with the M2-1 protein showing a greater partitioning into raft membranes compared to that of the N, P or M proteins. Determination of the polymerase activity profile of the gradient fractions revealed that 95% of the detectable viral enzyme activity was associated with lipid-raft membranes. Furthermore, analysis of virus-infected cells by confocal microscopy suggested an association between these proteins and the raft-lipid, GM1. Together, these results provide evidence that the RSV polymerase complex is able to associate with lipid rafts in virus-infected cells.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Membrane Microdomains/virology , Respiratory Syncytial Virus, Human/enzymology , Amino Acid Sequence , Cell Line, Tumor , DNA-Directed RNA Polymerases/chemistry , Humans , Mass Spectrometry , Molecular Sequence Data , Peptide Fragments/chemistry
12.
Virology ; 302(1): 207-16, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12429529

ABSTRACT

Clustered charge-to-alanine mutagenesis was performed on the large (L) polymerase protein of human respiratory syncytial virus to identify charged residues in the L protein that are important for viral RNA synthesis and to generate temperature-sensitive viruses. Clusters of three, four, and five charged residues throughout the entire L protein were substituted with alanines. A minigenome replicon assay was used to determine the functions of the mutant L proteins and to identify mutations that caused temperature sensitivity by comparing the level of reporter gene expression at 39 and 33 degrees C. Charge-to-alanine mutations were introduced into an antigenomic cDNA derived from RSV A2 strain to recover infectious viruses. Of the 27 charge-to-alanine mutations, 17 recombinant viruses (63%) were obtained. Seven mutants (41%) exhibited small plaque morphologies and/or temperature-sensitive growth in tissue culture. To generate mutant viruses with more temperature-sensitive and attenuated phenotypes, several clusters of charge-to-alanine substitutions were combined. Five combination mutants were recovered that exhibited shut-off temperatures ranging from 36 to 39 degrees C in tissue culture and restricted replication in the respiratory tracts of cotton rats.


Subject(s)
Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/metabolism , Alanine/genetics , Alanine/metabolism , Animals , Chlorocebus aethiops , Genome, Viral , Humans , Mutagenesis , Mutagenesis, Site-Directed , Phenotype , Rats , Recombination, Genetic , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology , Sigmodontinae , Temperature , Tumor Cells, Cultured , Vero Cells , Viral Proteins/genetics
13.
Nucleic Acids Res ; 26(23): 5327-32, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9826755

ABSTRACT

We have demonstrated that the human eosinophil-derived neurotoxin (EDN, RNase 2), a rapidly evolving secretory protein derived from eosinophilic leukocytes, mediates the ribonucleolytic destruction of extracellular virions of the single-stranded RNA virus respiratory syncytial virus (RSV). While RNase activity is crucial to antiviral activity, it is clearly not sufficient, as our results suggest that EDN has unique structural features apart from RNase activity that are necessary to promote antiviral activity. We demonstrate here that the interaction between EDN and extracellular virions of RSV is both saturatable and specific. Increasing concentrations of the antivirally inactivated, ribonucleolytically inactivated point mutant form of recombinant human EDN, rhEDNdK38, inhibits rhEDN's antiviral activity, while increasing concentrations of the related RNase, recombinant human RNase k6, have no effect whatsoever. Interestingly, acquisition of antiviral activity parallels the evolutionary development of the primate EDN lineage, having emerged some time after the divergence of the Old World from the New World monkeys. Using this information, we created ribonucleolytically active chimeras of human and New World monkey orthologs of EDN and, by evaluating their antiviral activity, we have identified an N-terminal segment of human EDN that contains one or more of the sequence elements that mediate its specific interaction with RSV.


Subject(s)
Antiviral Agents/pharmacology , Endoribonucleases/physiology , Evolution, Molecular , Proteins/physiology , Respiratory Syncytial Virus, Human/enzymology , Ribonuclease, Pancreatic/genetics , Ribonucleases , Amino Acid Sequence , Animals , Antiviral Agents/genetics , Antiviral Agents/physiology , Aotidae , Endoribonucleases/genetics , Eosinophil-Derived Neurotoxin , Extracellular Space/enzymology , Extracellular Space/virology , Humans , Macaca fascicularis , Molecular Sequence Data , Pongo pygmaeus , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Respiratory Syncytial Virus, Human/drug effects , Ribonuclease, Pancreatic/metabolism , Saguinus , Virion/physiology
14.
Virus Genes ; 13(3): 269-73, 1996.
Article in English | MEDLINE | ID: mdl-9035372

ABSTRACT

A cold-passaged (cp) temperature-sensitive (ts) mutant of human respiratory syncytial virus designated RSV cpts-248 was previously derived by random chemical mutagenesis of the non-ts mutant cp-RSV that possesses one or more host range mutations. We previously demonstrated in rodents and seronegative chimpanzees that the cpts-248 virus is more attenuated than cp-RSV and is more stable genetically than previously isolated RSV ts mutants. In the present study, we determined that the acquisition of the ts phenotype and the increased attenuation of the cpts-248 virus are associated with a single nucleotide substitution at nucleotide 10,989 that results in a change in the coding region (amino acid position 831) of the polymerase gene. The identification of this attenuating ts mutation is important because cpts-248 was used as the parent virus for the generation of a number of further attenuated mutants that are currently being evaluated as candidate vaccine strains in clinical trials in infants. Furthermore, technology now exists to rationally design new vaccine candidates by incorporating multiple attenuating mutations, such as the one identified here, into infectious viruses that are genetically stable and appropriately attenuated.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/genetics , Viral Vaccines/genetics , Animals , Base Sequence , Chlorocebus aethiops , Cold Temperature , DNA, Viral , Genes, Viral , Humans , Molecular Sequence Data , Mutagenesis , Phenotype , Respiratory Syncytial Virus, Human/genetics , Vero Cells
15.
J Gen Virol ; 75 ( Pt 3): 555-65, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8126452

ABSTRACT

P protein, the structural phosphoprotein of the Long strain of respiratory syncytial (RS) virus, is phosphorylated at serine residues. Some of these residues are candidates for modification by casein kinase II, as they are contained in consensus sequences. A cellular protein kinase, able to phosphorylate the P protein in vitro and apparently associated with purified RS virions, has been partially purified from HEp-2 cells. It shows several characteristics similar to those of casein kinase II. The P protein is modified in vitro by this activity mainly at serine residues located near the C terminus, which are also modified during virus infection. Thus, the P protein is phosphorylated in vivo in two regions, a central region as previously described, and another located in the C-terminal part of the molecule. The protein kinase involved in the phosphorylation of the C-terminal domain is similar to a cellular casein kinase II.


Subject(s)
HN Protein , Protein Kinases/metabolism , Respiratory Syncytial Virus, Human/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Protein Kinases/isolation & purification , Respiratory Syncytial Virus, Human/enzymology , Vero Cells , Viral Envelope Proteins , Virion/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...