1
|
Bonneux B, Jacoby E, Ceconi M, Stobbelaar K, Delputte P, Herschke F. Direct-acting antivirals for RSV treatment, a review. Antiviral Res 2024; 229:105948. [PMID: 38972604 DOI: 10.1016/j.antiviral.2024.105948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024]
Abstract
Respiratory syncytial virus (RSV) causes respiratory disease and complications in infants, the elderly and the immunocompromised. While three vaccines and two prophylactic monoclonal antibodies are now available, only one antiviral, ribavirin, is currently approved for treatment. This review aims to summarize the current state of treatments directly targeting RSV. Two major viral processes are attractive for RSV-specific antiviral drug discovery and development as they play essential roles in the viral cycle: the entry/fusion process carried out by the fusion protein and the replication/transcription process carried out by the polymerase complex constituted of the L, P, N and M2-1 proteins. For each viral target resistance mutations to small molecules of different chemotypes seem to delineate definite binding pockets in the fusion proteins and in the large proteins. Elucidating the mechanism of action of these inhibitors thus helps to understand how the fusion and polymerase complexes execute their functions. While many inhibitors have been studied, few are currently in clinical development for RSV treatment: one is in phase III, three in phase II and two in phase I. Progression was halted for many others because of strategic decisions, low enrollment, safety, but also lack of efficacy. Lessons can be learnt from the halted programs to increase the success rate of the treatments currently in development.
Collapse
Affiliation(s)
- Brecht Bonneux
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610, Wilrijk, Belgium; Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Edgar Jacoby
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Martina Ceconi
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610, Wilrijk, Belgium
| | - Kim Stobbelaar
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610, Wilrijk, Belgium.
| | | |
Collapse
|
2
|
Bivacqua R, Barreca M, Spanò V, Raimondi MV, Romeo I, Alcaro S, Andrei G, Barraja P, Montalbano A. Insight into non-nucleoside triazole-based systems as viral polymerases inhibitors. Eur J Med Chem 2023; 249:115136. [PMID: 36708678 DOI: 10.1016/j.ejmech.2023.115136] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Viruses have been recognized as the etiological agents responsible for many pathological conditions ranging from asymptomatic infections to serious diseases, even leading to death. For this reason, many efforts have been made to identify selective viral targets with the aim of developing efficient therapeutic strategies, devoid of drug-resistance issues. Considering their crucial role in the viral life cycle, polymerases are very attractive targets. Among the classes of compounds explored as viral polymerases inhibitors, here we present an overview of non-nucleoside triazole-based compounds identified in the last fifteen years. Furthermore, the structure-activity relationships (SAR) of the different chemical entities are described in order to highlight the key chemical features required for the development of effective antiviral agents.
Collapse
Affiliation(s)
- Roberta Bivacqua
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Marilia Barreca
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Maria Valeria Raimondi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy.
| | - Isabella Romeo
- Dipartimento di Scienze della Salute, Università Magna Græcia, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Academic Spinoff, Università Magna Græcia, Viale Europa, 88100, Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università Magna Græcia, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Academic Spinoff, Università Magna Græcia, Viale Europa, 88100, Catanzaro, Italy
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, 3000, Belgium
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| |
Collapse
|
3
|
Discovery of a Novel Respiratory Syncytial Virus Replication Inhibitor. Antimicrob Agents Chemother 2021; 65:AAC.02576-20. [PMID: 33782012 PMCID: PMC8316115 DOI: 10.1128/aac.02576-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/18/2021] [Indexed: 11/20/2022] Open
Abstract
A high-throughput screen of a Roche internal chemical library based on inhibition of the respiratory syncytial virus (RSV)-induced cytopathic effect (CPE) on HEp-2 cells was performed to identify RSV inhibitors. Over 2,000 hits were identified and confirmed to be efficacious against RSV infection in vitro Here, we report the discovery of a triazole-oxadiazole derivative, designated triazole-1, as an RSV replication inhibitor, and we characterize its mechanism of action. Triazole-1 inhibited the replication of both RSV A and B subtypes with 50% inhibitory concentration (IC50) values of approximately 1 μM, but it was not effective against other viruses, including influenza virus A, human enterovirus 71 (EV71), and vaccinia virus. Triazole-1 was shown to inhibit RSV replication when added at up to 8 h after viral entry, suggesting that it inhibits RSV after viral entry. In a minigenome reporter assay in which RSV transcription regulatory sequences flanking a luciferase gene were cotransfected with RSV N/P/L/M2-1 genes into HEp-2 cells, triazole-1 demonstrated specific and dose-dependent RSV transcription inhibitory effects. Consistent with these findings, deep sequencing of the genomes of triazole-1-resistant mutants revealed a single point mutation (A to G) at nucleotide 13546 of the RSV genome, leading to a T-to-A change at amino acid position 1684 of the L protein, which is the RSV RNA polymerase for both viral transcription and replication. The effect of triazole-1 on minigenome transcription, which was mediated by the L protein containing the T1684A mutation, was significantly reduced, suggesting that the T1684A mutation alone conferred viral resistance to triazole-1.
Collapse
|
4
|
Sutto-Ortiz P, Tcherniuk S, Ysebaert N, Abeywickrema P, Noël M, Decombe A, Debart F, Vasseur JJ, Canard B, Roymans D, Rigaux P, Eléouët JF, Decroly E. The methyltransferase domain of the Respiratory Syncytial Virus L protein catalyzes cap N7 and 2'-O-methylation. PLoS Pathog 2021; 17:e1009562. [PMID: 33956914 PMCID: PMC8130918 DOI: 10.1371/journal.ppat.1009562] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/18/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a negative sense single-stranded RNA virus and one of the main causes of severe lower respiratory tract infections in infants and young children. RSV RNA replication/transcription and capping are ensured by the viral Large (L) protein. The L protein contains a polymerase domain associated with a polyribonucleotidyl transferase domain in its N-terminus, and a methyltransferase (MTase) domain followed by the C-terminal domain (CTD) enriched in basic amino acids at its C-terminus. The MTase-CTD of Mononegavirales forms a clamp to accommodate RNA that is subsequently methylated on the cap structure and depending on the virus, on internal positions. These enzymatic activities are essential for efficient viral mRNA translation into proteins, and to prevent the recognition of uncapped viral RNA by innate immunity sensors. In this work, we demonstrated that the MTase-CTD of RSV, as well as the full-length L protein in complex with phosphoprotein (P), catalyzes the N7- and 2’-O-methylation of the cap structure of a short RNA sequence that corresponds to the 5’ end of viral mRNA. Using different experimental systems, we showed that the RSV MTase-CTD methylates the cap structure with a preference for N7-methylation as first reaction. However, we did not observe cap-independent internal methylation, as recently evidenced for the Ebola virus MTase. We also found that at μM concentrations, sinefungin, a S-adenosylmethionine analogue, inhibits the MTase activity of the RSV L protein and of the MTase-CTD domain. Altogether, these results suggest that the RSV MTase domain specifically recognizes viral RNA decorated by a cap structure and catalyzes its methylation, which is required for translation and innate immune system subversion. Respiratory syncytial virus (RSV) is responsible of infant bronchiolitis and severe lower respiratory tract infections in infants and young children, and the leading cause of hospitalization in children under one year of age. However, we still lack a vaccine and therapeutics against this important pathogen. The main enzymatic activities involved in RSV propagation are embedded in the Large (L) protein that contains the polymerase domain and also all the activities required for RNA cap structure synthesis and methylation. These post-transcriptional RNA modifications play a key role in virus replication because cap N7-methylation is required for viral RNA translation into proteins, and 2’-O-methylation hides viral RNA from innate immunity detection. Viral methyltransferase (MTase) activities are now considered potential antiviral targets because their inhibition might limit the virus production and strengthen early virus detection by innate immunity sensors. In this work, we compared the enzymatic activities of the MTase expressed as a single domain or in the context of the full-length L protein. We demonstrated that the MTase protein catalyzes the specific methylation of the cap structure at both N7- and 2’-O-positions, and we obtained the proof of concept that a S-adenosylmethionine analogue can inhibit the MTase activity of the L protein.
Collapse
Affiliation(s)
| | - Sergey Tcherniuk
- Unité de Virologie et Immunologie Moléculaires, INRAE, Université Paris Saclay, Jouy en Josas, France
| | - Nina Ysebaert
- Janssen Infectious Diseases and Vaccines, Beerse, Belgium
| | | | - Mathieu Noël
- IBMM, Université de Montpellier, ENSCM, CNRS, UMR 5247, Montpellier, France
| | - Alice Decombe
- Aix Marseille Université, CNRS, AFMB UMR 7257, Marseille, France
| | - Françoise Debart
- IBMM, Université de Montpellier, ENSCM, CNRS, UMR 5247, Montpellier, France
| | | | - Bruno Canard
- Aix Marseille Université, CNRS, AFMB UMR 7257, Marseille, France
| | - Dirk Roymans
- Janssen Infectious Diseases and Vaccines, Beerse, Belgium
| | - Peter Rigaux
- Janssen Infectious Diseases and Vaccines, Beerse, Belgium
| | - Jean-François Eléouët
- Unité de Virologie et Immunologie Moléculaires, INRAE, Université Paris Saclay, Jouy en Josas, France
| | - Etienne Decroly
- Aix Marseille Université, CNRS, AFMB UMR 7257, Marseille, France
- * E-mail:
| |
Collapse
|
5
|
Winkley K, Koseva B, Banerjee D, Cheung W, Selvarangan R, Pastinen T, Grundberg E. High-resolution epigenome analysis in nasal samples derived from children with respiratory viral infections reveals striking changes upon SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.09.21253155. [PMID: 33758880 PMCID: PMC7987039 DOI: 10.1101/2021.03.09.21253155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background DNA methylation patterns of the human genome can be modified by environmental stimuli and provide dense information on gene regulatory circuitries. We studied genome-wide DNA methylation in nasal samples from infants (<6 months) applying whole-genome bisulfite sequencing (WGBS) to characterize epigenome response to 10 different respiratory viral infections including SARS-CoV-2. Results We identified virus-specific differentially methylated regions (vDMR) with human metapneumovirus (hMPV) and SARS-CoV-2 followed by Influenza B (Flu B) causing the weakest vs. strongest epigenome response with 496 vs. 78541 and 14361 vDMR, respectively. We found a strong replication rate of FluB (52%) and SARS-CoV-2 (42%) vDMR in independent samples indicating robust epigenome perturbation upon infection. Among the FluB and SARS-CoV-2 vDMRs, around 70% were hypomethylated and significantly enriched among epithelial cell-specific regulatory elements whereas the hypermethylated vDMRs for these viruses mapped more frequently to immune cell regulatory elements, especially those of the myeloid lineage. The hypermethylated vDMRs were also enriched among genes and genetic loci in monocyte activation pathways and monocyte count. Finally, we perform single-cell RNA-sequencing characterization of nasal mucosa in response to these two viruses to functionally analyze the epigenome perturbations. Which supports the trends we identified in methylation data and highlights and important role for monocytes. Conclusions All together, we find evidence indicating genetic predisposition to innate immune response upon a respiratory viral infection. Our genome-wide monitoring of infant viral response provides first catalogue of associated host regulatory elements. Assessing epigenetic variation in individual patients may reveal evidence for viral triggers of childhood disease.
Collapse
Affiliation(s)
- Konner Winkley
- Department of Pediatrics, Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, Missouri, US
| | - Boryana Koseva
- Department of Pediatrics, Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, Missouri, US
| | - Dithi Banerjee
- Department of Pathology and Laboratory Medicine, Children’s Mercy Kansas City, Kansas City, Missouri, US
| | - Warren Cheung
- Department of Pediatrics, Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, Missouri, US
| | - Rangaraj Selvarangan
- Department of Pathology and Laboratory Medicine, Children’s Mercy Kansas City, Kansas City, Missouri, US
| | - Tomi Pastinen
- Department of Pediatrics, Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, Missouri, US
| | - Elin Grundberg
- Department of Pediatrics, Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, Missouri, US
| |
Collapse
|
6
|
In Vitro Primer-Based RNA Elongation and Promoter Fine Mapping of the Respiratory Syncytial Virus. J Virol 2020; 95:JVI.01897-20. [PMID: 33028717 PMCID: PMC7737744 DOI: 10.1128/jvi.01897-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a nonsegmented negative-sense (NNS) RNA virus and shares a similar RNA synthesis strategy with other members of NNS RNA viruses, such as measles, rabies virus, and Ebola virus. RSV RNA synthesis is catalyzed by a multifunctional RNA-dependent RNA polymerase (RdRP), which is composed of a large (L) protein that catalyzes three distinct enzymatic functions and an essential coenzyme phosphoprotein (P). Here, we successfully prepared highly pure, full-length, wild-type and mutant RSV polymerase (L-P) complexes. We demonstrated that the RSV polymerase could carry out both de novo and primer-based RNA synthesis. We defined the minimal length of the RNA template for in vitro de novo RNA synthesis using the purified RSV polymerase as 8 nucleotides (nt), shorter than previously reported. We showed that the RSV polymerase catalyzed primer-dependent RNA elongation with different lengths of primers on both short (10-nt) and long (25-nt) RNA templates. We compared the sequence specificity of different viral promoters and identified positions 3, 5, and 8 of the promoter sequence as essential to the in vitro RSV polymerase activity, consistent with the results previously mapped with the in vivo minigenome assay. Overall, these findings agree well with those of previous biochemical studies and extend our understanding of the promoter sequence and the mechanism of RSV RNA synthesis.IMPORTANCE As a major human pathogen, RSV affects 3.4 million children worldwide annually. However, no effective antivirals or vaccines are available. An in-depth mechanistic understanding of the RSV RNA synthesis machinery remains a high priority among the NNS RNA viruses. There is a strong public health need for research on this virus, due to major fundamental gaps in our understanding of NNS RNA virus replication. As the key enzyme executing transcription and replication of the virus, the RSV RdRP is a logical target for novel antiviral drugs. Therefore, exploring the primer-dependent RNA elongation extends our mechanistic understanding of the RSV RNA synthesis. Further fine mapping of the promoter sequence paves the way to better understand the function and structure of the RSV polymerase.
Collapse
|
7
|
Whelan JN, Reddy KD, Uversky VN, Teng MN. Functional correlations of respiratory syncytial virus proteins to intrinsic disorder. MOLECULAR BIOSYSTEMS 2017; 12:1507-26. [PMID: 27062995 DOI: 10.1039/c6mb00122j] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Protein intrinsic disorder is an important characteristic demonstrated by the absence of higher order structure, and is commonly detected in multifunctional proteins encoded by RNA viruses. Intrinsically disordered regions (IDRs) of proteins exhibit high flexibility and solvent accessibility, which permit several distinct protein functions, including but not limited to binding of multiple partners and accessibility for post-translational modifications. IDR-containing viral proteins can therefore execute various functional roles to enable productive viral replication. Respiratory syncytial virus (RSV) is a globally circulating, non-segmented, negative sense (NNS) RNA virus that causes severe lower respiratory infections. In this study, we performed a comprehensive evaluation of predicted intrinsic disorder of the RSV proteome to better understand the functional role of RSV protein IDRs. We included 27 RSV strains to sample major RSV subtypes and genotypes, as well as geographic and temporal isolate differences. Several types of disorder predictions were applied to the RSV proteome, including per-residue (PONDR®-FIT and PONDR® VL-XT), binary (CH, CDF, CH-CDF), and disorder-based interactions (ANCHOR and MoRFpred). We classified RSV IDRs by size, frequency and function. Finally, we determined the functional implications of RSV IDRs by mapping predicted IDRs to known functional domains of each protein. Identification of RSV IDRs within functional domains improves our understanding of RSV pathogenesis in addition to providing potential therapeutic targets. Furthermore, this approach can be applied to other NNS viruses that encode essential multifunctional proteins for the elucidation of viral protein regions that can be manipulated for attenuation of viral replication.
Collapse
Affiliation(s)
- Jillian N Whelan
- Division of Allergy and Immunology, Department of Internal Medicine, and the Joy McCann Culverhouse Airway Diseases Research Center, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Krishna D Reddy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA and Institute for Biological Instrumentation, Russian Academy of Sciences, 142292 Pushchino, Moscow Region, Russia
| | - Michael N Teng
- Division of Allergy and Immunology, Department of Internal Medicine, and the Joy McCann Culverhouse Airway Diseases Research Center, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
8
|
Koehler A, Kolesnikova L, Becker S. An active site mutation increases the polymerase activity of the guinea pig-lethal Marburg virus. J Gen Virol 2016; 97:2494-2500. [PMID: 27450090 DOI: 10.1099/jgv.0.000564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Marburg virus (MARV) causes severe, often fatal, disease in humans and transient illness in rodents. Sequential passaging of MARV in guinea pigs resulted in selection of a lethal virus containing 4 aa changes. A D184N mutation in VP40 (VP40D184N), which leads to a species-specific gain of viral fitness, and three mutations in the active site of viral RNA-dependent RNA polymerase L, which were investigated in the present study for functional significance in human and guinea pig cells. The transcription/replication activity of L mutants was strongly enhanced by a substitution at position 741 (S741C), and inhibited by other substitutions (D758A and A759D) in both species. The polymerase activity of L carrying the S741C substitution was eightfold higher in guinea pig cells than in human cells upon co-expression with VP40D184N, suggesting that the additive effect of the two mutations provides MARV a replicative advantage in the new host.
Collapse
Affiliation(s)
- Alexander Koehler
- Institut für Virologie, Philipps-Universität Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany
| | - Larissa Kolesnikova
- Institut für Virologie, Philipps-Universität Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany
| | - Stephan Becker
- German Center for Infection Research (DZIF), Institut für Virologie, Philipps-Universität, Hans-Meerwein-Str. 2, 35043 Marburg, Germany.,Institut für Virologie, Philipps-Universität Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany
| |
Collapse
|
9
|
Do LAH, Wilm A, van Doorn HR, Lam HM, Sim S, Sukumaran R, Tran AT, Nguyen BH, Tran TTL, Tran QH, Vo QB, Dac NAT, Trinh HN, Nguyen TTH, Binh BTL, Le K, Nguyen MT, Thai QT, Vo TV, Ngo NQM, Dang TKH, Cao NH, Tran TV, Ho LV, Farrar J, de Jong M, Chen S, Nagarajan N, Bryant JE, Hibberd ML. Direct whole-genome deep-sequencing of human respiratory syncytial virus A and B from Vietnamese children identifies distinct patterns of inter- and intra-host evolution. J Gen Virol 2016; 96:3470-3483. [PMID: 26407694 DOI: 10.1099/jgv.0.000298] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is the major cause of lower respiratory tract infections in children ,2 years of age. Little is known about RSV intra-host genetic diversity over the course of infection or about the immune pressures that drive RSV molecular evolution. We performed whole-genome deep-sequencing on 53 RSV-positive samples (37 RSV subgroup A and 16 RSV subgroup B) collected from the upper airways of hospitalized children in southern Vietnam over two consecutive seasons. RSV A NA1 and RSV B BA9 were the predominant genotypes found in our samples, consistent with other reports on global RSV circulation during the same period. For both RSV A and B, the M gene was the most conserved, confirming its potential as a target for novel therapeutics. The G gene was the most variable and was the only gene under detectable positive selection. Further, positively selected sites inG were found in close proximity to and in some cases overlapped with predicted glycosylation motifs, suggesting that selection on amino acid glycosylation may drive viral genetic diversity. We further identified hotspots and coldspots of intra-host genetic diversity in the RSV genome, some of which may highlight previously unknown regions of functional importance.
Collapse
Affiliation(s)
- Lien Anh Ha Do
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Ho Chi Minh City, Vietnam
| | - Andreas Wilm
- Genome Institute of Singapore, Genome Building, 138672 Singapore
| | - H Rogier van Doorn
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Ho Chi Minh City, Vietnam.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ha Minh Lam
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Ho Chi Minh City, Vietnam
| | - Shuzhen Sim
- Genome Institute of Singapore, Genome Building, 138672 Singapore
| | - Rashmi Sukumaran
- Genome Institute of Singapore, Genome Building, 138672 Singapore
| | - Anh Tuan Tran
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | - Bach Hue Nguyen
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | - Thi Thu Loan Tran
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | - Quynh Huong Tran
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | - Quoc Bao Vo
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | | | - Hong Nhien Trinh
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | | | - Bao Tinh Le Binh
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | - Khanh Le
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | - Minh Tien Nguyen
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | - Quang Tung Thai
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | - Thanh Vu Vo
- Children's Hospital 1, Ward 10, District 10, Ho Chi Minh City, Vietnam
| | | | - Thi Kim Huyen Dang
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | - Ngoc Huong Cao
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | - Thu Van Tran
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | - Lu Viet Ho
- Children's Hospital 2, Ben Nghe Ward, District 1, Ho Chi Minh City, Vietnam
| | - Jeremy Farrar
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Ho Chi Minh City, Vietnam
| | - Menno de Jong
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Ho Chi Minh City, Vietnam.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Swaine Chen
- Genome Institute of Singapore, Genome Building, 138672 Singapore
| | | | - Juliet E Bryant
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Ho Chi Minh City, Vietnam.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Martin L Hibberd
- Genome Institute of Singapore, Genome Building, 138672 Singapore
| |
Collapse
|
10
|
The Interferon Type I/III Response to Respiratory Syncytial Virus Infection in Airway Epithelial Cells Can Be Attenuated or Amplified by Antiviral Treatment. J Virol 2015; 90:1705-17. [PMID: 26608311 DOI: 10.1128/jvi.02417-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/16/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Human respiratory syncytial virus (RSV) is a single-stranded RNA virus that causes acute, and occasionally fatal, lower respiratory illness in young infants, the elderly, and immunocompromised patients. Therapeutic interventions able to cut short viral replication and quickly return the airways to normal function are needed. An understanding of antiviral activities and their effects on host defense mechanisms is important for the design of safe and effective therapy. We targeted functionally and temporally distinct steps within the viral life cycle using small-molecule RSV inhibitors and studied their antiviral activities and their effects on innate interferon responses of airway epithelial cells in vitro. Antivirals acting upstream of RSV polymerase activity (i.e., compounds targeting the fusion protein or the nucleoprotein) reduced viral load immediately postinfection and partially attenuated interferon responses. In contrast, antivirals directed to the RSV polymerase demonstrated activity throughout the viral replication cycle and specifically modulated the RIG-I/mitochondrial antiviral signaling protein (MAVS)/TBK1/IRF3/interferon-stimulated gene (ISG) axis, causing either an upregulation or a downregulation of interferon responses, depending on the mechanism of polymerase inhibition. Notably, polymerase inhibition leading to the accumulation of abortive RNA products correlated with the amplification of interferon-stimulated genes to up to 10 times above normal infection levels. Understanding how antiviral activities and their modulation of innate immunity may affect recovery from RSV infection will help guide the development of safe and effective therapies. IMPORTANCE RSV circulates seasonally, causing acute lower respiratory disease. Therapeutic interventions with efficacy throughout the viral replication cycle, rapid viral clearance, and prevention of potentially harmful inflammatory responses are desirable. Compounds targeting the RSV polymerase inhibited virus replication late in the viral life cycle and, depending on the functional domain targeted, either attenuated or amplified RIG-I and downstream interferon pathways in infected cells. These data will help guide the development of safe and effective therapies by providing new molecular evidence that the mechanism of inhibition by an antiviral compound can directly impact innate antiviral immune responses in the airway epithelium.
Collapse
|
11
|
Kim YI, DeVincenzo JP, Jones BG, Rudraraju R, Harrison L, Meyers R, Cehelsky J, Alvarez R, Hurwitz JL. Respiratory syncytial virus human experimental infection model: provenance, production, and sequence of low-passaged memphis-37 challenge virus. PLoS One 2014; 9:e113100. [PMID: 25415360 PMCID: PMC4240712 DOI: 10.1371/journal.pone.0113100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/19/2014] [Indexed: 01/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children and is responsible for as many as 199,000 childhood deaths annually worldwide. To support the development of viral therapeutics and vaccines for RSV, a human adult experimental infection model has been established. In this report, we describe the provenance and sequence of RSV Memphis-37, the low-passage clinical isolate used for the model's reproducible, safe, experimental infections of healthy, adult volunteers. The predicted amino acid sequences for major proteins of Memphis-37 are compared to nine other RSV A and B amino acid sequences to examine sites of vaccine, therapeutic, and pathophysiologic interest. Human T- cell epitope sequences previously defined by in vitro studies were observed to be closely matched between Memphis-37 and the laboratory strain RSV A2. Memphis-37 sequences provide baseline data with which to assess: (i) virus heterogeneity that may be evident following virus infection/transmission, (ii) the efficacy of candidate RSV vaccines and therapeutics in the experimental infection model, and (iii) the potential emergence of escape mutants as a consequence of experimental drug treatments. Memphis-37 is a valuable tool for pre-clinical research, and to expedite the clinical development of vaccines, therapeutic immunomodulatory agents, and other antiviral drug strategies for the protection of vulnerable populations against RSV disease.
Collapse
Affiliation(s)
- Young-In Kim
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Children's Foundation Research Institute of Le Bonheur Children's Hospital, Memphis, Tennessee, United States of America
| | - John P. DeVincenzo
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Children's Foundation Research Institute of Le Bonheur Children's Hospital, Memphis, Tennessee, United States of America
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Rajeev Rudraraju
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Lisa Harrison
- Children's Foundation Research Institute of Le Bonheur Children's Hospital, Memphis, Tennessee, United States of America
| | - Rachel Meyers
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Jeff Cehelsky
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Rene Alvarez
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Julia L. Hurwitz
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
12
|
Hiriote W, Gias ELM, Welsh SH, Toms GL. An investigation of the genetic basis of increased susceptibility to neutralization by anti-fusion glycoprotein antibody arising on passage of human respiratory syncytial virus in cell culture. J Med Virol 2014; 87:130-40. [PMID: 24861209 DOI: 10.1002/jmv.23980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2014] [Indexed: 11/09/2022]
Abstract
Human respiratory syncytial virus isolates have previously been shown to exhibit resistance to neutralization by anti-fusion glycoprotein antibodies that is lost on passage in cell culture. Early passage resistant and late passage susceptible stocks of two virus isolates from different epidemics were cloned by plaque purification. Early passage stocks of both isolates yielded predominantly neutralization resistant clones while late passage stocks yielded predominantly susceptible clones. On further characterization of resistant and susceptible clones, resistant virus yields were lower and they were relatively resistant to both neutralization and fusion inhibition by anti-F murine monoclonal antibodies and were also resistant to neutralization by human sera and by Palivizumab. The full genome of resistant and susceptible clones from one of the isolates was sequenced. Four differences, confirmed by sequencing sister clones, were found between resistant and susceptible clones, one in each of the SH, G, F, and L genes.
Collapse
Affiliation(s)
- W Hiriote
- The Institute of Cellular Medicine, The University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | | | | | | |
Collapse
|
13
|
Application of minigenome technology in virology research of the Paramyxoviridae family. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2014; 48:123-9. [PMID: 24767419 DOI: 10.1016/j.jmii.2014.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/12/2014] [Accepted: 02/19/2014] [Indexed: 11/23/2022]
Abstract
Minigenomes (MGs) are complementary DNAs of the synthetic analogs of genomic RNA. MGs are widely used to study the life cycle of the Paramyxoviridae family of viruses. MG-based studies have provided valuable insights into the mechanisms of viral replication and transcription in this family, including the roles of viral proteins, the location and boundaries of the cis-acting elements, the functional domains of trans-acting proteins, techniques for the measurement of neutralizing antibody, virus-host interactions, and the structure and function of viral RNA. This article provides a brief overview of the principle and application of MG technology in studies involving members of the Paramyxoviridae family. The advantages, potential limitations, and future scope of MG technology are also discussed.
Collapse
|
14
|
Hotard AL, Shaikh FY, Lee S, Yan D, Teng MN, Plemper RK, Crowe JE, Moore ML. A stabilized respiratory syncytial virus reverse genetics system amenable to recombination-mediated mutagenesis. Virology 2012; 434:129-36. [PMID: 23062737 PMCID: PMC3492879 DOI: 10.1016/j.virol.2012.09.022] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/06/2012] [Accepted: 09/21/2012] [Indexed: 10/27/2022]
Abstract
We describe the first example of combining bacterial artificial chromosome (BAC) recombination-mediated mutagenesis with reverse genetics for a negative strand RNA virus. A BAC-based respiratory syncytial virus (RSV) rescue system was established. An important advantage of this system is that RSV antigenomic cDNA was stabilized in the BAC vector. The RSV genotype chosen was A2-line19F, a chimeric strain previously shown to recapitulate in mice key features of RSV pathogenesis. We recovered two RSV reporter viruses, one expressing the red fluorescent protein monomeric Katushka 2 (A2-K-line19F) and one expressing Renilla luciferase (A2-RL-line19F). As proof of principle, we efficiently generated a RSV gene deletion mutant (A2-line19FΔNS1/NS2) and a point mutant (A2-K-line19F-I557V) by recombination-mediated BAC mutagenesis. Together with sequence-optimized helper expression plasmids, BAC-RSV is a stable, versatile, and efficient reverse genetics platform for generation of a recombinant Pneumovirus.
Collapse
Affiliation(s)
- Anne L. Hotard
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Fyza Y. Shaikh
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sujin Lee
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Dan Yan
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Michael N. Teng
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Richard K. Plemper
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - James E. Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Martin L. Moore
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
15
|
Abstract
Nipah (NiV) and Hendra (HeV) viruses comprise the genus Henipavirus and are highly pathogenic paramyxoviruses, which cause fatal encephalitis and respiratory disease in humans. Since their respective initial outbreaks in 1998 and 1994, they have continued to cause sporadic outbreaks resulting in fatal disease. Due to their designation as Biosafety Level 4 pathogens, the level of containment required to work with live henipaviruses is available only to select laboratories around the world. This chapter provides an overview of the molecular virology of NiV and HeV including comparisons to other, well-characterized paramyxoviruses. This chapter also describes the sequence diversity present among the henipaviruses.
Collapse
Affiliation(s)
- Paul A Rota
- MS-C-22, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | | |
Collapse
|
16
|
Ksiazek TG, Rota PA, Rollin PE. A review of Nipah and Hendra viruses with an historical aside. Virus Res 2011; 162:173-83. [PMID: 21963678 DOI: 10.1016/j.virusres.2011.09.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 09/17/2011] [Accepted: 09/18/2011] [Indexed: 11/29/2022]
Abstract
The emergence of Hendra and Nipah viruses in the 1990s has been followed by the further emergence of these viruses in the tropical Old World. The history and current knowledge of the disease, the viruses and their epidemiology is reviewed in this article. A historical aside summarizes the role that Dr. Brian W.J. Mahy played at critical junctures in the early stories of these viruses.
Collapse
Affiliation(s)
- Thomas G Ksiazek
- Galveston National Laboratory, Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0610, USA.
| | | | | |
Collapse
|
17
|
Fix J, Galloux M, Blondot ML, Eléouët JF. The insertion of fluorescent proteins in a variable region of respiratory syncytial virus L polymerase results in fluorescent and functional enzymes but with reduced activities. Open Virol J 2011; 5:103-8. [PMID: 21966341 PMCID: PMC3178903 DOI: 10.2174/1874357901105010103] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/29/2011] [Accepted: 07/13/2011] [Indexed: 11/22/2022] Open
Abstract
The respiratory syncytial virus (RSV) Large protein L is the catalytic subunit of the RNA-dependent RNA polymerase complex. Currently, no structural information is available for RSV L. Sequence alignments of L protein from human and bovine strains of RSV revealed the existence of two variable regions, VR1 and VR2. Following comparison with morbillivirus and rhabdovirus L genes, VR2, which is located between domains V and VI, was chosen as an insertion site for sequences encoding the epitope tag HA or the fluorescent proteins eGFP and mCherry. Recombinant tagged-L proteins co-localized with RSV N and P proteins in transfected cells. These recombinant polymerases were shown to be functional using a viral minigenome system assay, their activities being reduced by ~70% compared to the unmodified L polymerase. We have also shown by site-directed mutagenesis that the GDNQ motif (residues 810-813 for the Long strain of HRSV) is essential for L activity.
Collapse
Affiliation(s)
- Jenna Fix
- INRA, Unité de Virologie Immunologie Moléculaires UR892, F-78350 Jouy-en-Josas, France
| | | | | | | |
Collapse
|
18
|
Kumaria R, Iyer LR, Hibberd ML, Simões EAF, Sugrue RJ. Whole genome characterization of non-tissue culture adapted HRSV strains in severely infected children. Virol J 2011; 8:372. [PMID: 21794174 PMCID: PMC3166936 DOI: 10.1186/1743-422x-8-372] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 07/28/2011] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Human respiratory syncytial virus (HRSV) is the most important virus causing lower respiratory infection in young children. The complete genetic characterization of RSV clinical strains is a prerequisite for understanding HRSV infection in the clinical context. Current information about the genetic structure of the HRSV genome has largely been obtained using tissue culture adapted viruses. During tissue culture adaptation genetic changes can be introduced into the virus genome, which may obscure subtle variations in the genetic structure of different RSV strains. METHODS In this study we describe a novel Sanger sequencing strategy which allowed the complete genetic characterisation of 14 clinical HRSV strains. The viruses were sequenced directly in the nasal washes of severely hospitalized children, and without prior passage of the viruses in tissue culture. RESULTS The analysis of nucleotide sequences suggested that vRNA length is a variable factor among primary strains, while the phylogenetic analysis suggests selective pressure for change. The G gene showed the greatest sequence variation (2-6.4%), while small hydrophobic protein and matrix genes were completely conserved across all clinical strains studied. A number of sequence changes in the F, L, M2-1 and M2-2 genes were observed that have not been described in laboratory isolates. The gene junction regions showed more sequence variability, and in particular the intergenic regions showed a highest level of sequence variation. Although the clinical strains grew slower than the HRSVA2 virus isolate in tissue culture, the HRSVA2 isolate and clinical strains formed similar virus structures such as virus filaments and inclusion bodies in infected cells; supporting the clinical relevance of these virus structures. CONCLUSION This is the first report to describe the complete genetic characterization of HRSV clinical strains that have been sequenced directly from clinical material. The presence of novel substitutions and deletions in the vRNA of clinical strains emphasize the importance of genomic characterization of non-tissue culture adapted primary strains.
Collapse
Affiliation(s)
- Rajni Kumaria
- Singapore-MIT Alliance for Research and Technology, Centre for Life Sciences, #05-06M, 28 Medical Drive, 117456, Singapore
| | - Laxmi Ravi Iyer
- Division of Molecular and Cell biology, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 639798, Singapore
| | - Martin L Hibberd
- Singapore-MIT Alliance for Research and Technology, Centre for Life Sciences, #05-06M, 28 Medical Drive, 117456, Singapore
- Genome Institute of Singapore, #02-01, Genome Building, 60 Biopolis Street, 138672, Singapore
| | - Eric AF Simões
- University of Colorado, Denver and The Division of Infectious Diseases, The Children's Hospital, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | - Richard J Sugrue
- Singapore-MIT Alliance for Research and Technology, Centre for Life Sciences, #05-06M, 28 Medical Drive, 117456, Singapore
- Division of Molecular and Cell biology, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 639798, Singapore
| |
Collapse
|
19
|
Llorente MT, Taylor IA, López-Viñas E, Gomez-Puertas P, Calder LJ, García-Barreno B, Melero JA. Structural properties of the human respiratory syncytial virus P protein: evidence for an elongated homotetrameric molecule that is the smallest orthologue within the family of paramyxovirus polymerase cofactors. Proteins 2008; 72:946-58. [PMID: 18300250 DOI: 10.1002/prot.21988] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
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.
Collapse
Affiliation(s)
- María T Llorente
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
20
|
Lai SH, Stein DA, Guerrero-Plata A, Liao SL, Ivanciuc T, Hong C, Iversen PL, Casola A, Garofalo RP. Inhibition of respiratory syncytial virus infections with morpholino oligomers in cell cultures and in mice. Mol Ther 2008; 16:1120-8. [PMID: 18443602 PMCID: PMC2782410 DOI: 10.1038/mt.2008.81] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infection in infants, young children, and high-risk adults. Currently, there is no vaccine to prevent RSV infection, and the available therapeutic agents are of limited utility. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are a class of antisense agents that can enter cells readily and interfere with viral protein expression through steric blocking of complementary RNA. Two antisense PPMOs, designed to target sequence that includes the 5'-terminal region and translation start-site region of RSV L mRNA, were tested for anti-RSV activity in cultures of two human-airway cell lines. Both PPMOs showed minimal cytotoxicity and one of them, (AUG-2), reduced viral titers by >2.0 log(10). Intranasal (i.n.) treatment of BALB/c mice with AUG-2 PPMO before the RSV inoculation produced a reduction in viral titer of 1.2 log(10) in lung tissue at day 5 postinfection (p.i.), and attenuated pulmonary inflammation at day 7 postinfection. These data show that the AUG-2 PPMO possesses potent anti-RSV activity and is worthy of further investigation as a candidate for potential therapeutic application.
Collapse
Affiliation(s)
- Shen-Hao Lai
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Chang Gung Children’s Hospital, 5-7, Fu-Hsing Street, Kuei Shan Hsiang, Taoyuan Hsien, Taiwan, R.O.C., and Chang Gung University, 259, Wen Hwa 1 Rd., Taoyuan, Taiwan 33333
| | - David A. Stein
- AVI BioPharma Inc., 4575 SW Research Way, Corvallis OR, 97333, USA
| | - Antonieta Guerrero-Plata
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | - Sui-Ling Liao
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Chang Gung Children’s Hospital, 5-7, Fu-Hsing Street, Kuei Shan Hsiang, Taoyuan Hsien, Taiwan, R.O.C., and Chang Gung University, 259, Wen Hwa 1 Rd., Taoyuan, Taiwan 33333
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | - Chao Hong
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | | | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | - Roberto P. Garofalo
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| |
Collapse
|
21
|
Carromeu C, Simabuco FM, Tamura RE, Farinha Arcieri LE, Ventura AM. Intracellular localization of human respiratory syncytial virus L protein. Arch Virol 2007; 152:2259-63. [PMID: 17703289 DOI: 10.1007/s00705-007-1048-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 07/19/2007] [Indexed: 11/24/2022]
Abstract
Replication and transcription of the human respiratory syncytial virus (hRSV) genome is carried out by the ribonucleocapsid complex (RNA together with N, P, M2-1 and L proteins), with the L protein being responsible for all enzymatic activities. In the present study, we obtained anti-L polyclonal sera in mice. These antibodies were functional in immunofluorescence and Western blotting assays in hRSV-infected HEp-2 cells. In the immunofluorescence assays, we detected inclusion bodies in the anti-L staining, similar to the ones seen by anti-N or anti-P staining. The results presented here provide the first evidence of the intracellular localization of the hRSV L protein.
Collapse
Affiliation(s)
- C Carromeu
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
22
|
Abstract
Pneumovirus infection remains a significant problem for both human and veterinary medicine. Both avian pneumovirus (aMPV, Turkey rhinotracheitis virus) and human metapneumovirus (hMPV) are pathogens of birds and humans, which are associated with respiratory tract infections. Based on their different genomic organization and low level of nucleotide (nt) and amino acid (aa) identity with paramyxoviruses in the genus Pneumovirus, aMPV and hMPV have been classified into a new genus referred to as Metapneumovirus. The advancement of our understanding of pneumovirus biology and pathogenesis of pneumovirus disease in specific natural hosts can provide us with strategies for vaccine formulations and combined antiviral and immunomodulatory therapies.
Collapse
Affiliation(s)
- Shobha Broor
- Department of Microbiology, All India Institute of Medical Sciences Hospital, New Delhi, India.
| | | |
Collapse
|
23
|
Barik S, Bitko V. Prospects of RNA interference therapy in respiratory viral diseases: update 2006. Expert Opin Biol Ther 2006; 6:1151-60. [PMID: 17049013 DOI: 10.1517/14712598.6.11.1151] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Respiratory viruses, such as influenza, parainfluenza and respiratory syncytial virus (RSV), claim millions of lives annually. At present, there is no completely effective vaccine or drug against these highly mutable RNA viruses. Passive antibody therapies for RSV, despite their limited application and staggering cost, enjoy a virtual monopoly in a multibillion-dollar global market. Recently, however, pioneering discoveries have launched RNA interference as a novel, nucleic acid-based therapy against viral pathogens. Specifically, small interfering RNAs (siRNAs) offered protection against respiratory syncytial virus, parainfluenza and influenza. siRNA against RSV has entered Phase I clinical trials in humans, and preliminary reports are promising. If appropriately formulated for improved specificity, delivery and pharmacokinetics, siRNAs may indeed become effective antivirals in the clinics of the future. This paper provides an overview of the prospects and hurdles facing the antiviral siRNA drugs, with special emphasis on RSV.
Collapse
Affiliation(s)
- Sailen Barik
- University of South Alabama College of Medicine, Department of Biochemistry and Molecular Biology, 307 University Blvd, Mobile, Alabama 36688-0002, USA.
| | | |
Collapse
|
24
|
Cowton VM, McGivern DR, Fearns R. Unravelling the complexities of respiratory syncytial virus RNA synthesis. J Gen Virol 2006; 87:1805-1821. [PMID: 16760383 DOI: 10.1099/vir.0.81786-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is the leading cause of paediatric respiratory disease and is the focus of antiviral- and vaccine-development programmes. These goals have been aided by an understanding of the virus genome architecture and the mechanisms by which it is expressed and replicated. RSV is a member of the order Mononegavirales and, as such, has a genome consisting of a single strand of negative-sense RNA. At first glance, transcription and genome replication appear straightforward, requiring self-contained promoter regions at the 3' ends of the genome and antigenome RNAs, short cis-acting elements flanking each of the genes and one polymerase. However, from these minimal elements, the virus is able to generate an array of capped, methylated and polyadenylated mRNAs and encapsidated antigenome and genome RNAs, all in the appropriate ratios to facilitate virus replication. The apparent simplicity of genome expression and replication is a consequence of considerable complexity in the polymerase structure and its cognate cis-acting sequences; here, our understanding of mechanisms by which the RSV polymerase proteins interact with signals in the RNA template to produce different RNA products is reviewed.
Collapse
MESH Headings
- Base Sequence
- DNA-Directed RNA Polymerases/metabolism
- Genome, Viral
- Humans
- Models, Biological
- Molecular Sequence Data
- Mononegavirales/genetics
- Mononegavirales/physiology
- Nucleocapsid/biosynthesis
- Nucleocapsid/genetics
- RNA, Antisense/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Viral/biosynthesis
- RNA, Viral/genetics
- Respiratory Syncytial Virus, Human/genetics
- Respiratory Syncytial Virus, Human/pathogenicity
- Respiratory Syncytial Virus, Human/physiology
- Transcription, Genetic
- Viral Proteins/metabolism
- Virus Replication
Collapse
Affiliation(s)
- Vanessa M Cowton
- Division of Pathology and Neuroscience, University of Dundee Medical School, Dundee DD1 9SY, UK
| | - David R McGivern
- Division of Pathology and Neuroscience, University of Dundee Medical School, Dundee DD1 9SY, UK
| | - Rachel Fearns
- Division of Pathology and Neuroscience, University of Dundee Medical School, Dundee DD1 9SY, UK
| |
Collapse
|
25
|
Melero JA. Molecular Biology of Human Respiratory Syncytial Virus. RESPIRATORY SYNCYTIAL VIRUS 2006. [DOI: 10.1016/s0168-7069(06)14001-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
26
|
Muthuchelvan D, Sanyal A, Singh RP, Hemadri D, Sen A, Sreenivasa BP, Singh RK, Bandyopadhyay SK. Comparative sequence analysis of the large polymerase protein (L) gene of peste-des-petits ruminants (PPR) vaccine virus of Indian origin. Arch Virol 2005; 150:2467-81. [PMID: 16052284 DOI: 10.1007/s00705-005-0596-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Accepted: 05/27/2005] [Indexed: 10/25/2022]
Abstract
The complete nucleotide sequence of the large polymerase (L) protein of the peste-des-petits ruminants (PPR) vaccine virus (PPRV Sungri/96) belonging to the Asian lineage was determined. The gene was 6643 nucleotides in length from the gene-start to the gene-end and encoded a polypeptide of 2183 amino acids. The PPRV Sungri/96 has a nucleotide homology of 94.1% for PPRV Nigeria 75/1 to 64.4% for Canine distemper virus. At amino acid level PPRV Sungri/96 has an amino acid identity of 96.2% with PPRV Nigeria 75/1 and 70.4% to 74.8% with other morbilliviruses. All the established domains in L protein characteristic of paramyxoviruses were also found to be present in PPRV Sungri/96. Phylogenetic analysis of different L proteins of morbilliviruses revealed five well-defined clusters as observed previously. The 3' trailer sequence of PPRV Sungri/96 is of 37 nucleotides long which is very similar to that of other morbilliviruses. To the best of our knowledge this is the first report describing the polymerase gene sequence of PPRV Indian isolate.
Collapse
Affiliation(s)
- D Muthuchelvan
- Central Institute of Fisheries Technology, Cochin, India
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Grdzelishvili VZ, Smallwood S, Tower D, Hall RL, Hunt DM, Moyer SA. A single amino acid change in the L-polymerase protein of vesicular stomatitis virus completely abolishes viral mRNA cap methylation. J Virol 2005; 79:7327-37. [PMID: 15919887 PMCID: PMC1143665 DOI: 10.1128/jvi.79.12.7327-7337.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The vesicular stomatitis virus (VSV) RNA polymerase synthesizes viral mRNAs with 5'-cap structures methylated at the guanine-N7 and 2'-O-adenosine positions (7mGpppA(m)). Previously, our laboratory showed that a VSV host range (hr) and temperature-sensitive (ts) mutant, hr1, had a complete defect in mRNA cap methylation and that the wild-type L protein could complement the hr1 defect in vitro. Here, we sequenced the L, P, and N genes of mutant hr1 and found only two amino acid substitutions, both residing in the L-polymerase protein, which differentiate hr1 from its wild-type parent. These mutations (N505D and D1671V) were introduced separately and together into the L gene, and their effects on VSV in vitro transcription and in vivo chloramphenicol acetyltransferase minigenome replication were studied under conditions that are permissive and nonpermissive for hr1. Neither L mutation significantly affected viral RNA synthesis at 34 degrees C in permissive (BHK) and nonpermissive (HEp-2) cells, but D1671V reduced in vitro transcription and genome replication by about 50% at 40 degrees C in both cell lines. Recombinant VSV bearing each mutation were isolated, and the hr and ts phenotypes in infected cells were the result of a single D1671V substitution in the L protein. While the mutations did not significantly affect mRNA synthesis by purified viruses, 5'-cap analyses of product mRNAs clearly demonstrated that the D1671V mutation abrogated all methyltransferase activity. Sequence analysis suggests that an aspartic acid at amino acid 1671 is a critical residue within a putative conserved S-adenosyl-l-methionine-binding domain of the L protein.
Collapse
Affiliation(s)
- Valery Z Grdzelishvili
- University of Florida College of Medicine, Department of Molecular Genetics and Microbiology, 1600 S.W. Archer Road, P.O. Box 100266, Gainesville, FL 32610-0266, USA
| | | | | | | | | | | |
Collapse
|
28
|
Krempl CD, Lamirande EW, Collins PL. Complete sequence of the RNA genome of pneumonia virus of mice (PVM). Virus Genes 2005; 30:237-49. [PMID: 15744580 DOI: 10.1007/s11262-004-5631-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Accepted: 10/04/2004] [Indexed: 11/24/2022]
Abstract
Pneumonia virus of mice (PVM) is an enveloped RNA-containing virus of Family Paramyxoviridae. Sequences had been determined previously for a number of PVM genes, although these represented cloned cDNAs rather than consensus sequences. Sequences were not available for the 3' -leader and 5' -trailer regions that constitute the genome termini or for the large polymerase L gene that accounts for 43% of the genome. Also, the available sequences were from an attenuated variant of strain 15, whereas the present study analyzed the version of strain 15 that is available from the American Type Culture Collection (ATCC) and is highly virulent in mice. Analysis of unclosed RT-PCR products yielded a complete consensus sequence of 14,886 nt (GenBank accession number AY729016). Of the regions for which sequences had been previously reported for the non-pathogenic strain, there were 13 nucleotide differences and 10 amino acid differences compared to the present consensus sequence for the virulent isolate. The various genes of PVM shared 29-62% nucleotide sequence identity and 10-60% amino acid sequence identity with human or bovine respiratory syncytial virus (HRSV and BRSV), its closest relatives.
Collapse
Affiliation(s)
- Christine D Krempl
- Department of Virology, Institute for Medical Microbiology and Hygiene, Albert-Ludwigs University, Freiburg im Breisgau, Germany
| | | | | |
Collapse
|
29
|
Govindarajan D, Samal SK. Sequence analysis of the large polymerase (L) protein of the US strain of avian metapneumovirus indicates a close resemblance to that of the human metapneumovirus. Virus Res 2005; 105:59-66. [PMID: 15325081 DOI: 10.1016/j.virusres.2004.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Revised: 04/21/2004] [Accepted: 04/21/2004] [Indexed: 11/27/2022]
Abstract
The complete nucleotide sequence of the large polymerase (L) protein of the avian metapneumovirus subgroup C strain Colorado was determined. The L protein gene of avian pneumovirus Colorado isolate (APV-C) was 6173 nucleotides in length from the gene-start to the gene-end and encoded a polypeptide of 2005 amino acids in length. The length of the L protein of APV-C was exactly the same as that of human metapneumovirus (hMPV) and one amino acid longer than the L protein of APV subgroup A. The L protein of APV-C showed 80% amino acid identity with the L protein of hMPV, but only 64% amino acid identity with the L protein of APV-A. The nucleotide and deduced amino acid sequences were compared with the corresponding sequences of eleven other paramyxoviruses. All six domains characteristic of paramyxovirus L proteins were also observed in the L protein of APV-C. All the polymerase core motifs in domain III were conserved to nearly 100% in the metapneumoviruses. Similarly, the putative ATP-binding motif in domain VI was completely conserved among the metapneumoviruses and differed in length, by one intermediate residue, from other paramyxoviruses. Phylogenetic analysis of the different L proteins also revealed a closer relationship between APV-C and hMPV.
Collapse
Affiliation(s)
- Dhanasekaran Govindarajan
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, 8075 Greenmead Drive, College Park, Maryland 20742, USA
| | | |
Collapse
|
30
|
Thorpe LC, Easton AJ. Genome sequence of the non-pathogenic strain 15 of pneumonia virus of mice and comparison with the genome of the pathogenic strain J3666. J Gen Virol 2005; 86:159-169. [PMID: 15604443 DOI: 10.1099/vir.0.80315-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pneumonia virus of mice (PVM) is a member of the subfamily Pneumovirinae and is the closest known relative of respiratory syncytial virus. Both viruses cause pneumonia in their respective hosts. Here, the genome sequences of two strains of PVM, non-pathogenic strain 15 and pathogenic strain J3666, are reported. Comparison of the genome sequences revealed 59 nucleotide differences between the two strains, 37 of which were coding. The nucleotide differences were spread throughout the genome, affecting cis-acting regulatory regions and seven of the ten genes. Development of a reverse-genetics system for PVM should allow further elucidation of the functional importance of the genetic differences between the two strains identified here.
Collapse
Affiliation(s)
- L C Thorpe
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - A J Easton
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
31
|
Easton AJ, Domachowske JB, Rosenberg HF. Animal pneumoviruses: molecular genetics and pathogenesis. Clin Microbiol Rev 2004; 17:390-412. [PMID: 15084507 PMCID: PMC387412 DOI: 10.1128/cmr.17.2.390-412.2004] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pneumoviruses are single-stranded, negative-sense, nonsegmented RNA viruses of the family Paramyxoviridae, subfamily Pneumovirinae, and include pathogens that infect humans (respiratory syncytial virus and human metapneumovirus), domestic mammals (bovine, ovine, and caprine respiratory syncytial viruses), rodents (pneumonia virus of mice), and birds (avian metapneumovirus). Among the topics considered in this review are recent studies focused on the roles of the individual virus-encoded components in promoting virus replication as well as in altering and evading innate antiviral host defenses. Advances in the molecular technology of pneumoviruses and the emergence of recombinant pneumoviruses that are leading to improved virus-based vaccine formulations are also discussed. Since pneumovirus infection in natural hosts is associated with a profound inflammatory response that persists despite adequate antiviral therapy, we also review the recent experimental treatment strategies that have focused on combined antiviral, anti-inflammatory, and immunomodulatory approaches.
Collapse
|
32
|
Toquin D, de Boisseson C, Beven V, Senne DA, Eterradossi N. Subgroup C avian metapneumovirus (MPV) and the recently isolated human MPV exhibit a common organization but have extensive sequence divergence in their putative SH and G genes. J Gen Virol 2003; 84:2169-2178. [PMID: 12867649 DOI: 10.1099/vir.0.19043-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The genes encoding the putative small hydrophobic (SH), attachment (G) and polymerase (L) proteins of the Colorado isolate of subgroup C avian pneumovirus (APV) were entirely or partially sequenced. They all included metapneumovirus (MPV)-like gene start and gene end sequences. The deduced Colorado SH protein shared 26.9 and 21.7 % aa identity with its counterpart in human MPV (hMPV) and APV subgroup A, respectively, but its only significant aa similarities were to hMPV. Conserved features included a common hydrophobicity profile with an unique transmembrane domain and the conservation of most extracellular cysteine residues. The Colorado putative G gene encoded several ORFs, the longer of which encoded a 252 aa long type II glycoprotein with aa similarities to hMPV G only (20.6 % overall aa identity with seven conserved N-terminal residues). The putative Colorado G protein shared, at best, 21.0 % aa identity with its counterparts in the other APV subgroups and did not contain the extracellular cysteine residues and short aa stretch highly conserved in other APVs. The N-terminal end of the Colorado L protein exhibited 73.6 and 54.9 % aa identity with hMPV and APV subgroup A, respectively, with four aa blocks highly conserved among Pneumovirus: Phylogenetic analysis performed on the nt sequences confirmed that the L sequences from MPVs were genetically related, whereas analysis of the G sequences revealed that among MPVs, only APV subgroups A, B and D clustered together, independently of both the Colorado isolate and hMPV, which shared weak genetic relatedness at the G gene level.
Collapse
Affiliation(s)
- D Toquin
- French Agency for Food Safety (AFSSA), Avian and Rabbit Virology Immunology and Parasitology Unit (VIPAC), BP53, 22440 Ploufragan, France
| | - C de Boisseson
- Virus Genetics and Biosecurity Unit (UGVB), BP53, 22440 Ploufragan, France
| | - V Beven
- Virus Genetics and Biosecurity Unit (UGVB), BP53, 22440 Ploufragan, France
| | - D A Senne
- United States Department of Agriculture (USDA), National Veterinary Services Laboratories (NVSL), PO Box 844, Ames, IA 50010, USA
| | - N Eterradossi
- French Agency for Food Safety (AFSSA), Avian and Rabbit Virology Immunology and Parasitology Unit (VIPAC), BP53, 22440 Ploufragan, France
| |
Collapse
|
33
|
Cartee TL, Megaw AG, Oomens AGP, Wertz GW. Identification of a single amino acid change in the human respiratory syncytial virus L protein that affects transcriptional termination. J Virol 2003; 77:7352-60. [PMID: 12805433 PMCID: PMC164798 DOI: 10.1128/jvi.77.13.7352-7360.2003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infectious Human respiratory syncytial virus (HRSV) with an aberrant RNA synthesis pattern was recovered from a cDNA clone. The virus displayed increased levels of polycistronic readthrough mRNAs resulting from failure of the polymerase to terminate transcription efficiently at the gene ends. An asparagine (N) to aspartic acid (D) change at amino acid 1049 in the large (L) polymerase protein was found to be responsible for the readthrough phenotype. Virus encoding N at position 1049 displayed an RNA synthesis pattern indistinguishable from the A2 strain of HRSV. We compared the transcriptional activities of polymerases that encoded an N or D at position 1049 (L(1049N) or L(1049D)) by using dicistronic subgenomic replicons and found that the alteration of amino acid 1049 specifically affected transcriptional termination but had no effect on genome replication. L(1049N) recognized and terminated transcription at each of the naturally occurring gene junctions with differing efficiencies but at significantly higher efficiency than L(1049D). The abilities of the polymerases to recognize the cis-acting gene end signals required for termination were compared by examining the effect of single nucleotide changes at positions 1 to 8 of the M gene end and were found to be similar. This work identifies a single amino acid position in the polymerase that specifically affects the ability of the polymerase to terminate transcription, but which does not affect genome replication or interaction with the M2-1 protein. This work shows the critical importance of the previously defined cis-acting signals for termination, confirms differential termination at the varied gene junctions, and shows that the polymerase responds to the cis-acting sequences similarly, whether it has N or D at position 1049.
Collapse
Affiliation(s)
- Tara L Cartee
- Department of Microbiology, University of Alabama School of Medicine, Birmingham, Alabama 35294, USA
| | | | | | | |
Collapse
|
34
|
Skiadopoulos MH, Vogel L, Riggs JM, Surman SR, Collins PL, Murphy BR. The genome length of human parainfluenza virus type 2 follows the rule of six, and recombinant viruses recovered from non-polyhexameric-length antigenomic cDNAs contain a biased distribution of correcting mutations. J Virol 2003; 77:270-9. [PMID: 12477832 PMCID: PMC140631 DOI: 10.1128/jvi.77.1.270-279.2003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Members of the Paramyxovirinae subfamily of the Paramyxoviridae family of viruses have the unusual requirement that the nucleotide length of the viral genome must be an even multiple of six in order for efficient RNA replication, and hence virus replication, to occur. Human parainfluenza virus type 2 (HPIV2) is the only member of the genus that has been reported to have a genome length that is not an even multiple of six, and it has also been recovered from a full-length antigenomic-sense cDNA that did not conform to the "rule of six." To reexamine the issue of nucleotide length in natural isolates of HPIV2, a complete consensus genomic sequence was determined for three HPIV2 strains: Greer, Vanderbilt/1994 (V94), and Vanderbilt/1998. Each of these strains was found to have a genome length of 15,654 nucleotides (nt), thus conforming in each case to the rule of six. To directly examine the requirement that the genomic length of HPIV2 be an even multiple of six, we constructed six full-length antigenomic HPIV2/V94 cDNAs that deviated from a polyhexameric length by 0 to 5 nt. Recombinant HPIV2s were readily recovered from all of the cDNAs, including those that did not conform to the rule of six. One recombinant HPIV2 isolate was completely sequenced for each of the nonpolyhexameric antigenomic cDNAs. These were found to contain small nucleotide insertions or deletions that conferred polyhexameric length to the recovered genome. Interestingly, almost all of the length corrections occurred within the hemagglutinin-neuraminidase and large polymerase genes or the intervening intergenic region and thus were proximal to the insert that caused the deviation from the rule of six. These results demonstrate, in the context of complete infectious virus, that HPIV2 has a strong and seemingly absolute requirement for a polyhexameric genome.
Collapse
Affiliation(s)
- Mario H Skiadopoulos
- Respiratory Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Tang RS, Nguyen N, Zhou H, Jin H. Clustered charge-to-alanine mutagenesis of human respiratory syncytial virus L polymerase generates temperature-sensitive viruses. Virology 2002; 302:207-16. [PMID: 12429529 DOI: 10.1006/viro.2002.1596] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
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.
Collapse
|
36
|
Sasaya T, Ishikawa K, Koganezawa H. The nucleotide sequence of RNA1 of Lettuce big-vein virus, genus Varicosavirus, reveals its relation to nonsegmented negative-strand RNA viruses. Virology 2002; 297:289-97. [PMID: 12083827 DOI: 10.1006/viro.2002.1420] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The complete nucleotide sequence of RNA1 from Lettuce big-vein virus (LBVV), the type member of the genus Varicosavirus, was determined. LBVV RNA1 consists of 6797 nucleotides and contains one large ORF that encodes a large (L) protein of 2040 amino acids with a predicted M(r) of 232,092. Northern blot hybridization analysis indicated that the LBVV RNA1 is a negative-sense RNA. Database searches showed that the amino acid sequence of L protein is homologous to those of L polymerases of nonsegmented negative-strand RNA viruses. A cluster dendrogram derived from alignments of the LBVV L protein and the L polymerases indicated that the L protein is most closely related to the L polymerases of plant rhabdoviruses. Transcription termination/polyadenylation signal-like poly(U) tracts that resemble those in rhabdovirus and paramyxovirus RNAs were present upstream and downstream of the coding region. Although LBVV is related to rhabdoviruses, a key distinguishing feature is that the genome of LBVV is segmented. The results reemphasize the need to reconsider the taxonomic position of varicosaviruses.
Collapse
Affiliation(s)
- Takahide Sasaya
- National Agricultural Research Center for Western Region, Zentsuji Campus, Kagawa, Zentsuji, Japan.
| | | | | |
Collapse
|
37
|
van den Hoogen BG, Bestebroer TM, Osterhaus ADME, Fouchier RAM. Analysis of the genomic sequence of a human metapneumovirus. Virology 2002; 295:119-32. [PMID: 12033771 DOI: 10.1006/viro.2001.1355] [Citation(s) in RCA: 334] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently described the isolation of a novel paramyxovirus from children with respiratory tract disease in The Netherlands. Based on biological properties and limited sequence information the virus was provisionally classified as the first nonavian member of the Metapneumovirus genus and named human metapneumovirus (hMPV). This report describes the analysis of the sequences of all hMPV open reading frames (ORFs) and intergenic sequences as well as partial sequences of the genomic termini. The overall percentage of amino acid sequence identity between APV and hMPV N, P, M, F, M2-1, M2-2, and L ORFs was 56 to 88%. Some nucleotide sequence identity was also found between the noncoding regions of the APV and hMPV genomes. Although no discernible amino acid sequence identity was found between two of the ORFs of hMPV and ORFs of other paramyxoviruses, the amino acid content, hydrophilicity profiles, and location of these ORFs in the viral genome suggest that they represent SH and G proteins. The high percentage of sequence identity between APV and hMPV, their similar genomic organization (3'-N-P-M-F-M2-SH-G-L-5'), and phylogenetic analyses provide evidence for the proposed classification of hMPV as the first mammalian metapneumovirus.
Collapse
Affiliation(s)
- Bernadette G van den Hoogen
- Department of Virology, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 50, Rotterdam, 3015 GE, The Netherlands
| | | | | | | |
Collapse
|
38
|
Bujnicki JM, Rychlewski L. In silico identification, structure prediction and phylogenetic analysis of the 2'-O-ribose (cap 1) methyltransferase domain in the large structural protein of ssRNA negative-strand viruses. Protein Eng Des Sel 2002; 15:101-8. [PMID: 11917146 DOI: 10.1093/protein/15.2.101] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The Escherichia coli RrmJ gene product has recently been shown to be the 23S rRNA:U2552 specific 2'-O-ribose methyltransferase (MTase) (RrmJ). Its structure has been solved and refined to 1.5 A resolution, demonstrating conservation of the three-dimensional fold and key catalytic side chains with the vaccinia virus VP39 protein, which functions as an mRNA 5'm(7)G-cap-N-specific 2'-O-ribose MTase. Using the amino acid sequence of RrmJ as an initial probe in an iterative search of sequence databases, we identified a homologous domain in the sequence of the L protein of non-segmented, negative-sense, single-stranded RNA viruses. The plausibility of the prediction was confirmed by homology modeling and checking whether important residues at substrate/ligand-binding sites were conserved. The predicted structural compatibility and the conservation of the active site between the novel putative MTase domain and genuine 2'-O-ribose MTases, together with the available results of biochemical studies, strongly suggest that this domain is a 5'm(7)G-cap-N-specific 2'-O-ribose MTase (i.e. the cap 1 MTase). Evolutionary relationships between these proteins are also discussed.
Collapse
Affiliation(s)
- Janusz M Bujnicki
- Bioinformatics Laboratory, International Institute of Cell and Molecular Biology, ul. ks. Trojdena 4, 02-109 Warsaw, Poland.
| | | |
Collapse
|
39
|
Cartee TL, Wertz GW. Respiratory syncytial virus M2-1 protein requires phosphorylation for efficient function and binds viral RNA during infection. J Virol 2001; 75:12188-97. [PMID: 11711610 PMCID: PMC116116 DOI: 10.1128/jvi.75.24.12188-12197.2001] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The M2-1 protein of respiratory syncytial (RS) virus is a transcriptional processivity and antitermination factor. The M2-1 protein has a Cys3His1 zinc binding motif which is essential for function, is phosphorylated, and has been shown to interact with the RS virus nucleocapsid (N) protein. In the work reported here, we determined the sites at which the M2-1 protein was phosphorylated and investigated the importance of these phosphorylated residues for M2-1 function in transcription. By combining protease digestion, matrix-assisted laser desorption ionization-time of flight mass spectrometry, and site-directed mutagenesis, we identified the phosphorylated residues as serines 58 and 61, not threonine 56 and serine 58 as previously reported. Serines 58 and 61 and the surrounding amino acids are in a consensus sequence for phosphorylation by casein kinase I. Consistent with this, we showed that the unphosphorylated M2-1 protein synthesized in Escherichia coli could be phosphorylated in vitro by casein kinase I. The effect of eliminating phosphorylation by site-specific mutagenesis of serines 58 and 61 on the function of the M2-1 protein in transcription of RS virus subgenomic replicons was assayed. The activities of the M2-1 protein phosphorylation mutants in transcriptional antitermination were tested over a range of concentrations and were found to be substantially inhibited at all concentrations. The data show that phosphorylation is important for the M2-1 protein function in transcription. However, mutation of the M2-1 phosphorylation sites did not interfere with the ability of the M2-1 protein to interact with the N protein in transfected cells. The interaction of the M2-1 and N proteins in cotransfected cells was found to be sensitive to RNase A, indicating that the M2-1-N protein interaction was mediated via RNA. Furthermore, the M2-1 protein was shown to bind monocistronic and polycistronic RS virus mRNAs during infection.
Collapse
Affiliation(s)
- T L Cartee
- Department of Microbiology, University of Alabama School of Medicine, Birmingham, Alabama 35294, USA
| | | |
Collapse
|
40
|
Haller AA, MacPhail M, Mitiku M, Tang RS. A single amino acid substitution in the viral polymerase creates a temperature-sensitive and attenuated recombinant bovine parainfluenza virus type 3. Virology 2001; 288:342-50. [PMID: 11601905 DOI: 10.1006/viro.2001.1106] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bovine parainfluenza virus type 3 (bPIV3) is under development as a live virus vaccine vector. The RNA genome of a recombinant bPIV3 harbored four nucleotide changes, one of which resulted in a mutation of the viral polymerase (A. A. Haller et al., 2000, J. Virol. 74, 11626-11635). The contribution of this conservative amino acid substitution (I1103V) in the polymerase to the temperature-sensitive and attenuation phenotypes of r-bPIV3 was investigated by creating a new virus, r-bPIV3(I), that expressed the wild-type polymerase. r-bPIV3(I) was not temperature-sensitive for growth in vitro and the replication of r-bPIV3(I) was no longer restricted in hamsters. The effect of the amino acid substitution in the polymerase was also studied in a chimeric bovine/human PIV3, a virus that displayed temperature-sensitive and attenuated phenotypes (A. A. Haller et al., 2000, J. Virol. 74, 11626-11635). It was not clear whether these defects were due to the impaired polymerase or the replacement of the bPIV3 surface glycoproteins with those of hPIV3. The results showed that the altered polymerase was indeed responsible for the temperature-sensitive phenotype of bovine/human PIV3 but did not appear to play a role in the attenuation phenotype.
Collapse
Affiliation(s)
- A A Haller
- Aviron, 297 North Bernardo Avenue, Mountain View, California 94043, USA.
| | | | | | | |
Collapse
|
41
|
Crowe JE, Firestone CY, Whitehead SS, Collins PL, Murphy BR. Acquisition of the ts phenotype by a chemically mutagenized cold-passaged human respiratory syncytial virus vaccine candidate results from the acquisition of a single mutation in the polymerase (L) gene. Virus Genes 2001; 13:269-73. [PMID: 9035372 DOI: 10.1007/bf00366988] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
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.
Collapse
Affiliation(s)
- J E Crowe
- Respiratory Viruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892-0720, USA
| | | | | | | | | |
Collapse
|
42
|
Haller AA, Miller T, Mitiku M, Coelingh K. Expression of the surface glycoproteins of human parainfluenza virus type 3 by bovine parainfluenza virus type 3, a novel attenuated virus vaccine vector. J Virol 2000; 74:11626-35. [PMID: 11090161 PMCID: PMC112444 DOI: 10.1128/jvi.74.24.11626-11635.2000] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bovine parainfluenza virus type 3 (bPIV3) is being evaluated as an intranasal vaccine for protection against human PIV3 (hPIV3). In young infants, the bPIV3 vaccine appears to be infectious, attenuated, immunogenic, and genetically stable, which are desirable characteristics for an RNA virus vector. To test the potential of the bPIV3 vaccine strain as a vector, an infectious DNA clone of bPIV3 was assembled and recombinant bPIV3 (r-bPIV3) was rescued. r-bPIV3 displayed a temperature-sensitive phenotype for growth in tissue culture at 39 degrees C and was attenuated in the lungs of Syrian golden hamsters. In order to test whether r-bPIV3 could serve as a vector, the fusion and hemagglutinin-neuraminidase genes of bPIV3 were replaced with those of hPIV3. The resulting bovine/human PIV3 was temperature sensitive for growth in Vero cells at 37 degrees C. The replication of bovine/human PIV3 was also restricted in the lungs of hamsters, albeit not as severely as was observed for r-bPIV3. Despite the attenuation phenotypes observed for r-bPIV3 and bovine/human PIV3, both of these viruses protected hamsters completely upon challenge with hPIV3. In summary, bPIV3 was shown to function as a virus vector that may be especially suitable for vaccination of infants and children against PIV3 and other viruses.
Collapse
Affiliation(s)
- A A Haller
- Aviron, Mountain View, California 94043, USA.
| | | | | | | |
Collapse
|
43
|
Hardy RW, Wertz GW. The Cys(3)-His(1) motif of the respiratory syncytial virus M2-1 protein is essential for protein function. J Virol 2000; 74:5880-5. [PMID: 10846068 PMCID: PMC112083 DOI: 10.1128/jvi.74.13.5880-5885.2000] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The M2 gene of respiratory syncytial (RS) virus has two open reading frames (ORFs). ORF1 encodes a 22-kDa protein termed M2-1. The M2-1 protein contains a Cys(3)-His(1) motif (C-X(7)-C-X(5)-C-X(3)-H) near the amino terminus. This motif is conserved in all human, bovine, and ovine strains of RS virus. A similar motif found in the mammalian transcription factor Nup475 has been shown to bind zinc. The M2-1 protein of human RS virus functions as a transcription factor which increases polymerase processivity, and it enhances readthrough of intergenic junctions during RS virus transcription, thereby acting as a transcription antiterminator. The M2-1 protein also interacts with the nucleocapsid protein. We examined the effects of mutations of cysteine and histidine residues predicted to coordinate zinc in the Cys(3)-His(1) motif on transcription antitermination and N protein binding. We found that mutating the predicted zinc-coordinating residues, the cysteine residues at amino acid positions 7 and 15 and the histidine residue at position 25, prevented M2-1 from enhancing transcriptional readthrough. In contrast, mutations of amino acids within this motif not predicted to coordinate zinc had no effect. Mutations of the predicted zinc-coordinating residues in the Cys(3)-His(1) motif also prevented M2-1 from interacting with the nucleocapsid protein. One mutation of a noncoordinating residue in the motif which did not affect readthrough during transcription, E10G, prevented interaction with the nucleocapsid protein. This suggests that M2-1 does not require interaction with the nucleocapsid protein in order to function during transcription. Analysis of the M2-1 protein in reducing sodium dodecyl sulfate-polyacrylamide gels revealed two major forms distinguished by their mobilities. The slower migrating form was shown to be phosphorylated, whereas the faster migrating form was not. Mutations in the Cys(3)-His(1) motif caused a change in distribution of the M2-1 protein from the slower to the faster migrating form. The data presented here show that the Cys(3)-His(1) motif of M2-1 is essential for maintaining the functional integrity of the protein.
Collapse
Affiliation(s)
- R W Hardy
- Department of Microbiology, University of Alabama School of Medicine at Birmingham, 35294, USA
| | | |
Collapse
|
44
|
Respiratory syncytial virus: recent progress towards the discovery of effective prophylactic and therapeutic agents. Drug Discov Today 2000; 5:241-252. [PMID: 10825730 DOI: 10.1016/s1359-6446(00)01500-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Although respiratory syncytial virus (RSV) was discovered in 1955, the burden associated with this infectious agent on all population groups is only now beginning to be fully appreciated. The successful launch of the humanized monoclonal antibody Synagis (developed by MedImmune, Gaithersburg, MD, USA), as a prophylactic in September 1998 has helped to heighten awareness of the extent of mortality and morbidity associated with annual RSV epidemics. Small, drug-like molecules that would provide the clinician with effective and conveniently administered prophylactic and therapeutic agents for the prevention and treatment of RSV have not yet advanced into clinical studies. This review will summarize recent developments in the area of RSV drug discovery and development.
Collapse
|
45
|
Ahmadian G, Randhawa JS, Easton AJ. Expression of the ORF-2 protein of the human respiratory syncytial virus M2 gene is initiated by a ribosomal termination-dependent reinitiation mechanism. EMBO J 2000; 19:2681-9. [PMID: 10835365 PMCID: PMC212764 DOI: 10.1093/emboj/19.11.2681] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2000] [Revised: 04/10/2000] [Accepted: 04/10/2000] [Indexed: 11/13/2022] Open
Abstract
Translation of the open reading frame 2 (ORF-2) of the human respiratory syncytial virus M2 gene initiates at one of the three initiation codons located upstream of the termination codon for the first ORF. Replacement of ORF-2 with the major ORF of the chloramphenicol acetyltransferase reporter gene followed by systematic mutagenesis of the putative initiation codons demonstrated the usage of these codons as the translational initiators for ORF-2 expression both in vitro and in vivo. While the efficiency of translation was maintained when only the first and second AUG codons were preserved in vivo, there was no apparent preference in vitro for any of the three codons when only one was present. Mutagenesis studies showed that the location of the termination codon of ORF-1 protein plays a crucial role in directing translation of ORF-2 from the upstream initiation codons in vivo. This indicates that the second ORF is accessed by the ribosomes that are departing from the first ORF and that these ribosomes reinitiate on AUG codons 5' to the point of translation termination.
Collapse
Affiliation(s)
- G Ahmadian
- Department of Biological Sciences, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | | | | |
Collapse
|
46
|
Ahmadian G, Chambers P, Easton AJ. Detection and characterization of proteins encoded by the second ORF of the M2 gene of pneumoviruses. J Gen Virol 1999; 80 ( Pt 8):2011-2016. [PMID: 10466798 DOI: 10.1099/0022-1317-80-8-2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The nucleotide sequence of the M2 gene of pneumonia virus of mice (PVM) was determined. The sequence showed that the gene encoded a protein of 176 amino acids with a predicted molecular mass of 20165 Da from a major ORF, which is smaller than the equivalent proteins encoded by human, bovine and ovine respiratory syncytial (RS) viruses. The PVM M2 protein is conserved, having 41% similarity to the equivalent human RS virus protein. In common with the M2 genes of the RS viruses and avian pneumovirus (APV), the PVM mRNA also contained a second ORF (ORF2) that partially overlaps the first ORF and which is capable of encoding a 98 residue polypeptide. No significant sequence identity could be detected between the putative M2 ORF2 proteins of PVM, APV and the RS viruses. The expression of the M2 ORF2 proteins of the pneumoviruses was investigated by using monospecific antisera raised against GST fusion proteins. Western blot analysis demonstrated the presence of polypeptides encoded by M2 ORF2 of PVM and RS virus corresponding with those predicted by in vitro translation studies, but this was not the case for APV. The PVM polypeptide was present as three distinct products in vivo. The PVM and RS virus polypeptides were also detected in cells by immunofluorescence, which showed that both were present in the cytoplasm with a degree of localization in inclusion bodies. No APV M2 ORF2 protein could be detected in vivo. The RS virus M2 ORF2 polypeptide was shown to accumulate during infection and the potential implications of this are discussed.
Collapse
Affiliation(s)
- G Ahmadian
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK1
| | - P Chambers
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK1
| | - A J Easton
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK1
| |
Collapse
|
47
|
Skiadopoulos MH, Surman SR, St Claire M, Elkins WR, Collins PL, Murphy BR. Attenuation of the recombinant human parainfluenza virus type 3 cp45 candidate vaccine virus is augmented by importation of the respiratory syncytial virus cpts530 L polymerase mutation. Virology 1999; 260:125-35. [PMID: 10405364 DOI: 10.1006/viro.1999.9802] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A phenylalanine to leucine mutation at position 521 in the L polymerase of cpts530, a live-attenuated respiratory syncytial virus (RSV) cold-passaged (cp), temperature-sensitive (ts) candidate vaccine, specifies the ts and attenuation (att) phenotypes. Sequence alignment of this region in the L proteins of several distantly related paramyxoviruses revealed that this phenylalanine is conserved. Using reverse genetics, the analogous phenylalanine at position 456 in the L protein of wild-type PIV3 was mutagenized to leucine (F456L). The resulting virus, designated r456(L), was ts (40 degrees C shut-off temperature of plaque formation), and its replication in the upper, but not the lower, respiratory tract of hamsters was 10-fold reduced compared with that of the recombinant wild-type PIV3 (rwt). Thus the phenylalanine to leucine mutation specified a similar level of temperature sensitivity and attenuation in two distantly related paramyxoviruses. We next sought to determine whether the addition of this mutation to the L protein of two rPIV3 candidate vaccine viruses, one bearing the three cp45 ts missense mutations in the L protein (rcp45(L)) and the other bearing all 15 cp45 mutations (rcp45), would further attenuate the viruses in vivo. Each rcp45 derivative to which the F456L mutation was added exhibited an increased level of temperature sensitivity. Furthermore rcp45(L)-456 and rcp45-456 were 100- to 1000-fold more restricted in replication in hamsters than their rcp45(L) and rcp45 parents. Despite the high level of restriction of replication in hamsters, immunization with rcp45-456 induced a moderate level of resistance to replication of PIV3 challenge virus. In contrast to the highly restricted replication observed in hamsters, rcp45-456 was only fivefold more restricted in the respiratory tract of chimpanzees than rcp45 and induced a comparable, moderate to high level of PIV3-specific serum antibodies. rcp45 and rcp45-456 viruses isolated from chimpanzees throughout the 2-week course of replication maintained the level of temperature sensitivity of their respective input viruses, illustrating their phenotypic stability. Thus the acquisition of the F456L mutation by the cp45 virus resulted in a small, incremental increase in its level of attenuation, indicating its possible usefulness in the fine tuning of the level of attenuation of the cp45 vaccine candidate. The ability to transfer mutations identified in heterologous paramyxoviruses, which in this case represent different subfamilies, greatly enhances our ability to rapidly develop novel parainfluenza virus candidate vaccines.
Collapse
Affiliation(s)
- M H Skiadopoulos
- Laboratory of Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892-0720, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Marriott AC, Wilson SD, Randhawa JS, Easton AJ. A single amino acid substitution in the phosphoprotein of respiratory syncytial virus confers thermosensitivity in a reconstituted RNA polymerase system. J Virol 1999; 73:5162-5. [PMID: 10233981 PMCID: PMC112563 DOI: 10.1128/jvi.73.6.5162-5165.1999] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The single amino acid change Gly172 to Ser in the phosphoprotein (P) of respiratory syncytial virus (RSV) has previously been shown to be responsible for the thermosensitivity and protein-negative phenotype of tsN19, a mutant of the B subgroup RSN-2 strain. This single change was inserted into the P gene of the A subgroup virus RSS-2, and the resulting phenotype was observed in a plasmid-driven reconstituted RSV RNA polymerase system. Expression from a genome analogue containing two reporter genes was thermosensitive when directed by plasmids containing the N, L, M2, and mutant P genes cloned under the control of T7 promoters. Analysis of RNA synthesis showed that mutant P protein was unable to produce genome, antigenome, or mRNA at the restrictive temperature. At a semipermissive temperature, genome, antigenome, and mRNA synthesis were all reduced, 6- to 30-fold, relative to synthesis directed by a wild-type P plasmid. Binding of the mutant P protein to N protein in the absence of other viral proteins was unaffected by temperature, indicating that the lesion did not produce a large enough structural change to disrupt this binding. These data suggest that the plasmid rescue system is suitable for investigation of the role of thermosensitive mutations in RSV polymerase components in RNA synthesis.
Collapse
Affiliation(s)
- A C Marriott
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | | | | | | |
Collapse
|
49
|
Hardy RW, Harmon SB, Wertz GW. Diverse gene junctions of respiratory syncytial virus modulate the efficiency of transcription termination and respond differently to M2-mediated antitermination. J Virol 1999; 73:170-6. [PMID: 9847319 PMCID: PMC103820 DOI: 10.1128/jvi.73.1.170-176.1999] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/1998] [Accepted: 09/23/1998] [Indexed: 11/20/2022] Open
Abstract
The ability of the diverse gene junctions of respiratory syncytial (RS) virus to signal the termination of transcription was analyzed. Nine dicistronic subgenomic replicons of RS virus were constructed; each contained one of the RS virus gene junctions in its natural upstream and downstream sequence context. The RNA synthesis activities of these subgenomic replicons were analyzed in the absence and presence of the M2 protein, which we showed previously to function as a transcription antiterminator. Our data showed that the efficiency with which the polymerase terminated transcription was affected by the gene junction that it encountered. The M2 protein significantly decreased the efficiency of the termination of transcription, resulting in increased levels of readthrough transcription at all the gene junctions. The diverse gene junctions fell into three broad groups with respect to their ability to signal transcription termination. One group of gene junctions (NS1/NS2, NS2/N, M2/L, and L/trailer) showed inefficient termination in the absence or the presence of the M2 protein. A second group of gene junctions (N/P, P/M, M/SH, SH/G, and G/F) terminated transcription efficiently. The SH/G gene junction terminated transcription with the greatest efficiency and produced low levels of readthrough transcripts in the absence or the presence of the M2 protein, correlating with the absence of SH/G polycistronic transcripts in RS virus-infected cells. The F/M2 gene junction was particularly sensitive to the M2 protein: it efficiently signaled termination in the absence of the M2 protein but produced high levels of readthrough transcripts in the presence of the M2 protein. This result suggests that the M2 protein may regulate its own production by negative feedback. The data presented here show that the different gene junctions of RS virus do modulate RS virus transcription termination. The M2 protein reduced termination at all gene junctions. The magnitude of antitermination due to the M2 protein, however, varied at the different gene junctions. The data presented here indicate that the mechanism for the regulation of RS virus gene expression is more complex than was previously appreciated.
Collapse
Affiliation(s)
- R W Hardy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | |
Collapse
|
50
|
Buchholz UJ, Finke S, Conzelmann KK. Generation of bovine respiratory syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus replication in tissue culture, and the human RSV leader region acts as a functional BRSV genome promoter. J Virol 1999; 73:251-9. [PMID: 9847328 PMCID: PMC103829 DOI: 10.1128/jvi.73.1.251-259.1999] [Citation(s) in RCA: 844] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In order to generate recombinant bovine respiratory syncytial virus (BRSV), the genome of BRSV strain A51908, variant ATue51908, was cloned as cDNA. We provide here the sequence of the BRSV genome ends and of the entire L gene. This completes the sequence of the BRSV genome, which comprises a total of 15,140 nucleotides. To establish a vaccinia virus-free recovery system, a BHK-derived cell line stably expressing T7 RNA polymerase was generated (BSR T7/5). Recombinant BRSV was reproducibly recovered from cDNA constructs after T7 RNA polymerase-driven expression of antigenome sense RNA and of BRSV N, P, M2, and L proteins from transfected plasmids. Chimeric viruses in which the BRSV leader region was replaced by the human respiratory syncytial virus (HRSV) leader region replicated in cell culture as efficiently as their nonchimeric counterparts, demonstrating that all cis-acting sequences of the HRSV promoter are faithfully recognized by the BRSV polymerase complex. In addition, we report the successful recovery of a BRSV mutant lacking the complete NS2 gene, which encodes a nonstructural protein of unknown function. The NS2-deficient BRSV replicated autonomously and could be passaged, demonstrating that NS2 is not essential for virus replication in cell culture. However, growth of the mutant was considerably slower than and final infectious titers were reduced by a factor of at least 10 compared to wild-type BRSV, indicating that NS2 provides a supporting factor required for full replication capacity.
Collapse
Affiliation(s)
- U J Buchholz
- Department of Clinical Virology, Federal Research Center for Virus Diseases of Animals, D-72076 Tübingen, Germany
| | | | | |
Collapse
|