126 results on '"L protein"'
Search Results
2. Development of reverse genetics system for Guanarito virus: substitution of E1497K in the L protein of Guanarito virus S-26764 strain changes plaque phenotype and growth kinetics.
- Author
-
Satoshi Taniguchi, Maruyama, Junki, Takeshi Saito, Littlefield, Kirsten, Reyna, Rachel A., Manning, John T., Cheng Huang, Masayuki Saijo, and Paessler, Slobodan
- Subjects
- *
REVERSE genetics , *VIRAL proteins , *PHENOTYPIC plasticity , *GINGIVAL hemorrhage , *RNA polymerases , *CUCUMBER mosaic virus , *SUMATRIPTAN - Abstract
Guanarito virus (GTOV) is the causative agent of Venezuelan hemorrhagic fever. GTOV belongs to the genus Mammarenavirus, family Arenaviridae and has been classified as a Category A bioterrorism agent by the United States Centers for Disease Control and Prevention. Despite being a high-priority agent, vaccines and drugs against Venezuelan hemorrhagic fever are not available. GTOV S-26764, isolated from a nonfatal human case, produces an unclear cytopathic effect (CPE) in Vero cells, posing a significant obstacle to research and countermeasure development efforts. Vero celladapted GTOV S-26764 generated in this study produced clear CPE and demonstrated rapid growth and high yield in Vero cells compared to the original GTOV S-26764. We developed a reverse genetics system for GTOV to study amino acid changes acquired through Vero cell adaptation and leading to virus phenotype changes. The results demonstrated that E1497K in the L protein was responsible for the production of clear plaques as well as enhanced viral RNA replication and transcription efficiency. Vero cell-adapted GTOV S-26764, capable of generating CPE, will allow researchers to easily perform neutralization assays and anti-drug screening against GTOV. Moreover, the developed reverse genetics system will accelerate vaccine and antiviral drug development. IMPORTANCE Guanarito virus (GTOV) is a rodent-borne virus. GTOV causes fever, prostration, headache, arthralgia, cough, sore throat, nausea, vomiting, diarrhea, epistaxis, bleeding gums, menorrhagia, and melena in humans. The lethality rate is 23.1% or higher. Vero cell-adapted GTOV S-26764 shows a clear cytopathic effect (CPE), whereas the parental virus shows unclear CPE in Vero cells. We generated a reverse genetics system to rescue recombinant GTOVs and found that E1497K in the L protein was responsible for the formation of clear plaques as well as enhanced viral RNA replication and transcription efficiency. This reverse genetic system will accelerate vaccine and antiviral drug developments, and the findings of this study contribute to the understanding of the function of GTOV L as an RNA polymerase. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. HSP90 is part of a protein complex with the L polymerase of Rift Valley fever phlebovirus and prevents its degradation by the proteasome during the viral genome replication/transcription stage
- Author
-
Farhang Alem, Ashwini Brahms, Kaori Tarasaki, Samson Omole, Kylene Kehn-Hall, Connie S. Schmaljohn, Sina Bavari, Shinji Makino, and Ramin M. Hakami
- Subjects
Rift Valley fever virus ,RNA-dependent RNA polymerase ,L protein ,Hsp90 ,heat shock protein ,protein stability ,Microbiology ,QR1-502 - Abstract
The mosquito-borne Rift Valley fever virus (RVFV) from the Phenuiviridae family is a single-stranded RNA virus that causes the re-emerging zoonotic disease Rift Valley fever (RVF). Classified as a Category A agent by the NIH, RVFV infection can cause debilitating disease or death in humans and lead to devastating economic impacts by causing abortion storms in pregnant cattle. In a previous study, we showed that the host chaperone protein HSP90 is an RVFV-associated host factor that plays a critical role post viral entry, during the active phase of viral genome replication/transcription. In this study, we have elucidated the molecular mechanisms behind the regulatory effect of HSP90 during infection with RVFV. Our results demonstrate that during the early infection phase, host HSP90 associates with the viral RNA-dependent RNA polymerase (L protein) and prevents its degradation through the proteasome, resulting in increased viral replication.
- Published
- 2024
- Full Text
- View/download PDF
4. Discontinuous L-binding motifs in the transactivation domain of the vesicular stomatitis virus P protein are required for terminal de novo transcription initiation by the L protein.
- Author
-
Gupta, Nirmala, Minako Ogino, Watkins, Dean E., Tiffany Yu, Green, Todd J., and Tomoaki Ogino
- Subjects
- *
VESICULAR stomatitis , *VIRAL proteins , *AMINO acid residues , *MUTANT proteins , *ESSENTIAL amino acids , *RNA polymerases - Abstract
The phospho- (P) protein, the co-factor of the RNA polymerase large (L) protein, of vesicular stomatitis virus (VSV, a prototype of nonsegmented negative-strand RNA viruses) plays pivotal roles in transcription and replication. However, the precise mechanism underlying the transcriptional transactivation by the P protein has remained elusive. Here, using an in vitro transcription system and a series of deletion mutants of the P protein, we mapped a region encompassing residues 51-104 as a transactivation domain (TAD) that is critical for terminal de novo initiation, the initial step of synthesis of the leader RNA and anti-genome/genome, with the L protein. Site-directed mutagenesis revealed that conserved amino acid residues in three discontinuous L-binding sites within the TAD are essential for the transactivation activity of the P protein or important for maintaining its full activity. Importantly, relative inhibitory effects of TAD point mutations on synthesis of the full-length leader RNA and mRNAs from the 3'-terminal leader region and internal genes, respectively, of the genome were similar to those on terminal de novo initiation. Furthermore, any of the examined TAD mutations did not alter the gradient pattern of mRNAs synthesized from internal genes, nor did they induce the production of readthrough transcripts. These results suggest that these TAD mutations impact mainly terminal de novo initiation but rarely other steps (e.g., elongation, termination, internal initiation) of single-entry stop-start transcription. Consistently, the mutations of the essential or important amino acid residues within the P TAD were lethal or deleterious to VSV replication in host cells. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Understanding and Engineering Glycine Cleavage System and Related Metabolic Pathways for C1-Based Biosynthesis
- Author
-
Ren, Jie, Wang, Wei, Nie, Jinglei, Yuan, Wenqiao, Zeng, An-Ping, Scheper, Thomas, Series Editor, Belkin, Shimshon, Editorial Board Member, Bley, Thomas, Editorial Board Member, Bohlmann, Jörg, Editorial Board Member, Gu, Man Bock, Editorial Board Member, Hu, Wei Shou, Editorial Board Member, Mattiasson, Bo, Editorial Board Member, Olsson, Lisbeth, Editorial Board Member, Seitz, Harald, Editorial Board Member, Silva, Ana Catarina, Editorial Board Member, Ulber, Roland, Series Editor, Zeng, An-Ping, Editorial Board Member, Zhong, Jian-Jiang, Editorial Board Member, Zhou, Weichang, Editorial Board Member, and Claassens, Nico J., editor
- Published
- 2022
- Full Text
- View/download PDF
6. Genetic basis underlying Lassa fever endemics in the Mano River region, West Africa.
- Author
-
Li, Yan
- Subjects
- *
LASSA fever , *HEMORRHAGIC fever , *RNA replicase , *VIRAL proteins , *ENDEMIC diseases - Abstract
Lassa fever (LF), a haemorrhagic fever disease caused by Lassa virus (LASV), is a serious public health burden in West Africa. The Mano River region (Sierra Leone, Guinea, Liberia, and Côte d'Ivoire) has been an endemic focus of the disease over the past decades. Here, we deciphered the genetic basis underlying LF endemics in this region. Clade model and type I functional divergence analyses revealed that the major LASV group, Kenema sub-clade, which is currently circulating in the Eastern Province of Sierra Leone, has been affected by different selective pressure compared to isolates from the other areas with effects on the viral RNA-dependent RNA polymerase (L protein) and probably nucleoprotein (NP). Further, contingency analysis showed that, in the early endemic, the sub-clade has undergone adaptive diversification via acceleration of amino acid substitutions in L protein. These findings highlight the key viral factor and local adaptation regarding the endemicity of LF. • A molecular study was conducted on LF endemics in the Mano River region. • The major viral group, Kenema sub-clade, has been under local selection. • Viral L protein and probably NP have been the targets of the local selection. • In the early endemic, the Kenema sub-clade has experienced local adaptation. • The amino acid changes through the adaptation have accumulated on L protein. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Catalysis of mRNA Capping with GDP Polyribonucleotidyltransferase Activity of Rabies Virus L Protein
- Author
-
Ogino, Tomoaki, Green, Todd J., and Ahmad, Shamim I., editor
- Published
- 2021
- Full Text
- View/download PDF
8. Genomic profiling of three progenies isolated from a swarm of eGFP-tagged canine distemper viruses undergoing 40 serial passages.
- Author
-
Shuning Zhou, Ling Wang, Jiahui Lin, Ning Wang, Yongle Yu, Youming Zhang, and Fuxiao Liu
- Abstract
Canine distemper virus (CDV) is classified into the genus Morbillivirus in the family Paramyxoviridae. This virus has a single-stranded genomic RNA with negative polarity. The L protein is the viral RNA-dependent RNA polymerase (RdRp), closely associated with the fidelity of genome replication. We previously constructed an enhanced green fluorescence protein (eGFP)-tagged recombinant CDV (rCDV-eGFP). This recombinant underwent a total of 47 passages in vitro including 7 blind and 40 ribavirin-treated passages, consequently showing a rich diversity of viral quasispecies by the analysis of next-generation sequencing (NGS). In the present study, through one round of plaque purification, three single-plaque-derived variants (SPDVs) with the brightest fluorescence were independently isolated from a swarm of rCDV-eGFP progenies at passage-47, and then subjected to NGS analysis. The NGS results comprehensively uncovered mutation profiles of these three SPDVs. More importantly, this study aimed to unravel the potential relation between viral replication fidelity and L protein sequence. However, two SPDVs showed no single-amino acid mutation in their own L proteins, implying the rCDV-eGFP progenitor that might be a high-fidelity strain. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. CK1 and PP1 regulate Rift Valley fever virus genome replication through L protein phosphorylation.
- Author
-
Bracci, Nicole, Baer, Alan, Flor, Rafaela, Petraccione, Kaylee, Stocker, Timothy, Zhou, Weidong, Ammosova, Tatiana, Dinglasan, Rhoel R., Nekhai, Sergei, and Kehn-Hall, Kylene
- Subjects
- *
RIFT Valley fever , *RNA replicase , *VIRAL replication , *CASEIN kinase , *PHOSPHORYLATION - Abstract
Rift Valley fever virus (RVFV) is an arbovirus in the Phenuiviridae family identified initially by the large 'abortion storms' observed among ruminants; RVFV can also infect humans. In humans, there is a wide variation of clinical symptoms ranging from subclinical to mild febrile illness to hepatitis, retinitis, delayed-onset encephalitis, or even hemorrhagic fever. The RVFV is a tri-segmented negative-sense RNA virus consisting of S, M, and L segments. The L segment encodes the RNA-dependent RNA polymerase (RdRp), termed the L protein, which is responsible for both viral mRNA synthesis and genome replication. Phosphorylation of viral RdRps is known to regulate viral replication. This study shows that RVFV L protein is serine phosphorylated and identified Casein Kinase 1 alpha (CK1α) and protein phosphatase 1 alpha (PP1α) as L protein binding partners. Inhibition of CK1 and PP1 through small molecule inhibitor treatment, D4476 and 1E7-03, respectively, caused a change in the phosphorylated status of the L protein. Inhibition of PP1α resulted in increased L protein phosphorylation whereas inhibition of CK1α decreased L protein phosphorylation. It was also found that in RVFV infected cells, PP1α localized to the cytoplasmic compartment. Treatment of RVFV infected cells with CK1 inhibitors reduced virus production in both mammalian and mosquito cells. Lastly, inhibition of either CK1 or PP1 reduced viral genomic RNA levels. These data indicate that L protein is phosphorylated and that CK1 and PP1 play a crucial role in regulating the L protein phosphorylation cycle, which is critical to viral RNA production and viral replication. • RVFV L protein was found to be serine phosphorylated. • CK1α and PP1α are RVFV L protein interacting partners. • Inhibition of CK1 decreased viral titers in both mammalian and mosquito cells. • Inhibition of either PP1 or CK1 decreased viral genomic RNA levels. • CK1 and PP1 regulate the L protein phosphorylation cycle, which is critical to viral RNA production and viral replication. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Mutation Profiles of eGFP-Tagged Small Ruminant Morbillivirus During 45 Serial Passages in Ribavirin-Treated Cells
- Author
-
Fuxiao Liu, Yanli Zou, Lin Li, Chunju Liu, and Xiaodong Wu
- Subjects
rSRMV-eGFP ,next-generation sequencing ,single-nucleotide variation ,mutation ,L protein ,high-fidelity ,Veterinary medicine ,SF600-1100 - Abstract
Small ruminant morbillivirus (SRMV), formerly known as peste-des-petits-ruminants virus, classified into the genus Morbillivirus in the family Paramyxoviridae. Its L protein functions as the RNA-dependent RNA polymerases (RdRp) during viral replication. Due to the absence of efficient proofreading activity in their RdRps, various RNA viruses reveal high mutation frequencies, making them evolve rapidly during serial passages in cells, especially treated with a certain mutagen, like ribavirin. We have previously rescued a recombinant enhanced green fluorescence protein-tagged SRMV (rSRMV-eGFP) using reverse genetics. In this study, the rSRMV-eGFP was subjected to serial passages in ribavirin-treated cells. Due to the ribavirin-exerted selective pressure, it was speculated that viral progenies would form quasispecies after dozens of passages. Viral progenies at passage-10, -20, -30, -40, and -50 were separately subjected to next-generation sequencing (NGS), consequently revealing a total of 34 single-nucleotide variations, including five synonymous, 21 missense, and one non-sense mutations. The L sequence was found to harbor eight missense mutations during serial passaging. It was speculated that at least one high-fidelity variant was present in viral quasispecies at passage-50. If purified from the population of viral progenies, this putative variant would contribute to clarifying a molecular mechanism in viral high-fidelity replication in vitro.
- Published
- 2021
- Full Text
- View/download PDF
11. Hsp90 Is Required for Snakehead Vesiculovirus Replication via Stabilization of the Viral L Protein.
- Author
-
Yanwei Zhang, Yong-An Zhang, and Jiagang Tu
- Subjects
- *
VIRAL proteins , *SNAKEHEADS (Fish) , *FISH farming , *RNA synthesis - Abstract
Snakehead vesiculovirus (SHVV), a kind of fish rhabdovirus isolated from diseased hybrid snakehead fish, has caused great economic losses in snakehead fish culture in China. The large (L) protein, together with its cofactor phosphoprotein (P), forms a P/L polymerase complex and catalyzes the transcription and replication of viral genomic RNA. In this study, the cellular heat shock protein 90 (Hsp90) was identified as an interacting partner of SHVV L protein. Hsp90 activity was required for the stability of SHVV L because Hsp90 dysfunction caused by using its inhibitor destabilized SHVV L and thereby suppressed SHVV replication via reducing viral RNA synthesis. SHVV L expressed alone was detected mainly in the insoluble fraction, and the insoluble L was degraded by Hsp90 dysfunction through the proteasomal pathway, while the presence of SHVV P promoted the solubility of SHVV L and the soluble L was degraded by Hsp90 dysfunction through the autophagy pathway. Collectively, our data suggest that Hsp90 contributes to the maturation of SHVV L and ensures the effective replication of SHVV, which exhibits an important anti-SHVV target. This study will help us to understand the role of Hsp90 in stabilizing the L protein and regulating the replication of negative-stranded RNA viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. A substitution in the pre-S1 promoter region is associated with the viral regulation of hepatitis B virus
- Author
-
Suguru Ogura, Masahiko Tameda, Kazushi Sugimoto, Makoto Ikejiri, Masanobu Usui, Masaaki Ito, and Yoshiyuki Takei
- Subjects
Hepatitis B virus ,Mutation ,Pre-S1 promoter ,L protein ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Much evidence has demonstrated the influence of Hepatitis B virus (HBV) mutations on the clinical course of HBV infection. As large (L) protein plays a crucial role for viral entry, we hypothesized that mutations in the pre-S1 promoter region might affect the expression of L protein and subsequently change the biological characters of virus. Methods Patients infected with genotype C HBV were enrolled for analysis. HBV DNA sequences were inserted into a TA cloning vector and analyzed. To evaluate the effects of mutations in the pre-S1 promoter region, promoter activity and the expression of mRNA and L protein were analyzed using HepG2 cells. Results In total, 35 patients were enrolled and 13 patients (37.1%) had a single base substitution in the pre-S1 promoter region; the most frequent substitution was a G-to-A substitution at the 2765th base (G2765A) in the Sp1 region. The HBV viral load showed a negative correlation with the substitution ratio of the Sp1 region or G2765A (r = − 0.493 and − 0.473, respectively). Among those with a viral load ≤5.0 log IU/ml, patients with the G2765A substitution showed a significantly lower HBV viral load than those with the wild-type sequence. HepG2 cells transfected with the G2765A substitution vector showed reduced luciferase activity of the pre-S1 promoter, as well as reduced expression of pre-S1 mRNA and L protein. Furthermore, the G2765A substitution greatly reduced the L protein expression level of vector-produced virus particles. Conclusion G2765A substitution in the pre-S1 promoter reduced the expression of L protein and resulted in a low viral load and less severe disease in chronic HBV infections.
- Published
- 2019
- Full Text
- View/download PDF
13. Adaptive genetic diversification of Lassa virus associated with the epidemic split of north-central Nigerian and non-Nigerian lineages.
- Author
-
Li, Yan and Tian, Haifeng
- Subjects
- *
RNA replicase , *VIRAL proteins , *HEMORRHAGIC fever , *ENDEMIC diseases , *LASSA fever , *MOLECULAR clock - Abstract
Lassa fever (LF) is a viral hemorrhagic fever that causes high morbidity and severe mortality annually. The disease is endemic to two geographically separate areas within tropical West Africa, one in Nigeria and the second predominantly in Sierra Leone-Guinea-Liberia-Mali. Lassa virus (LASV), the causative agent of the disease, exhibits clear delineation of phylogeography between the endemic areas. In order to characterize the genetic nature of Nigerian–non-Nigerian epidemic split, we performed molecular epidemiological analyses on non-Nigerian isolates (lineage IV as well as lineage V) and their sister group from north-central Nigeria (lineage III). The results showed that adaptive genetic diversification has occurred between these currently circulating clusters in the spread process, and a number of replacement divergences have been fixed between these clusters on the viral RNA-dependent RNA polymerase (L protein). This study highlights the viral L protein could be a determinant factor for the epidemic split. • A genomic epidemiological study was conducted on LASV Nigerian–non-Nigerian split. • Adaptive selection has influenced the Nigerian–non-Nigerian epidemic split of LASV. • The adaptive genetic diversification has occurred on viral protein L. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. Development of reverse genetics system for Guanarito virus: substitution of E1497K in the L protein of Guanarito virus S-26764 strain changes plaque phenotype and growth kinetics.
- Author
-
Taniguchi S, Maruyama J, Saito T, Littlefield K, Reyna RA, Manning JT, Huang C, Saijo M, and Paessler S
- Subjects
- Animals, Female, Humans, Arenaviridae Infections virology, Arenaviruses, New World genetics, Chlorocebus aethiops, Hemorrhagic Fevers, Viral virology, Phenotype, Vaccines, Vero Cells, Arenaviridae genetics, Reverse Genetics methods
- Abstract
Guanarito virus (GTOV) is the causative agent of Venezuelan hemorrhagic fever. GTOV belongs to the genus Mammarenavirus , family Arenaviridae and has been classified as a Category A bioterrorism agent by the United States Centers for Disease Control and Prevention. Despite being a high-priority agent, vaccines and drugs against Venezuelan hemorrhagic fever are not available. GTOV S-26764, isolated from a non-fatal human case, produces an unclear cytopathic effect (CPE) in Vero cells, posing a significant obstacle to research and countermeasure development efforts. Vero cell-adapted GTOV S-26764 generated in this study produced clear CPE and demonstrated rapid growth and high yield in Vero cells compared to the original GTOV S-26764. We developed a reverse genetics system for GTOV to study amino acid changes acquired through Vero cell adaptation and leading to virus phenotype changes. The results demonstrated that E1497K in the L protein was responsible for the production of clear plaques as well as enhanced viral RNA replication and transcription efficiency. Vero cell-adapted GTOV S-26764, capable of generating CPE, will allow researchers to easily perform neutralization assays and anti-drug screening against GTOV. Moreover, the developed reverse genetics system will accelerate vaccine and antiviral drug development.IMPORTANCEGuanarito virus (GTOV) is a rodent-borne virus. GTOV causes fever, prostration, headache, arthralgia, cough, sore throat, nausea, vomiting, diarrhea, epistaxis, bleeding gums, menorrhagia, and melena in humans. The lethality rate is 23.1% or higher. Vero cell-adapted GTOV S-26764 shows a clear cytopathic effect (CPE), whereas the parental virus shows unclear CPE in Vero cells. We generated a reverse genetics system to rescue recombinant GTOVs and found that E1497K in the L protein was responsible for the formation of clear plaques as well as enhanced viral RNA replication and transcription efficiency. This reverse genetic system will accelerate vaccine and antiviral drug developments, and the findings of this study contribute to the understanding of the function of GTOV L as an RNA polymerase., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
15. L protein characterization and in silico screening of putative broad range target molecules for pathogenic mammarenaviruses from South America.
- Author
-
Rodrigues Dutra JV, Santos IA, Grosche VR, Jardim ACG, de Aguiar RS, Junior NN, and José DP
- Subjects
- South America, Molecular Docking Simulation, Arenaviridae drug effects, Computer Simulation, Amino Acid Sequence, Humans, Molecular Dynamics Simulation, Protein Binding, Phylogeny, Models, Molecular, Antiviral Agents pharmacology, Antiviral Agents chemistry, Viral Proteins chemistry, Viral Proteins antagonists & inhibitors, Viral Proteins metabolism
- Abstract
The genus Mammarenavirus belonging to the family Arenaviridae encompasses pathogenic viral species capable of triggering severe diseases in humans, causing concern for the health system due to the high fatality rate associated with them. Currently, there is a dearth of specific therapies against pathogens of the genus. Natural products isolated from plants have impacted the development of drugs against several diseases. The Núcleo de Bioensaios, Biossíntese e Ecofisiologia de Produtos Naturais (NuBBE) database offers several natural compounds with antimicrobial activities that can be used in the development of new antiviral drugs. In this context, here we modeled the arenavirus L protein, multifunctional machinery essential for the viral replicative cycle, making this enzyme a potential candidate for targeting the development of antivirals against genus pathogens. Using the modeled L protein, a virtual screening was performed, which suggested eleven molecules from the NuBBE database that binds to the active site of the L protein, which was promising in the in silico predictions of absorption and toxicity analysis. The NuBBE 1642 molecule proved to be the best candidate for four of the five species evaluated, acting as a possible broad-spectrum molecule. Additionally, our results showed that the L protein is highly conserved among species of the genus, as well as presenting close phylogenetic relationships between many of the species studied, strengthening its candidacy as a therapeutic target. The data presented here demonstrate that some NuBBE molecules are potential ligands for the L protein of arenaviruses, which may help to contain possible outbreaks.Communicated by Ramaswamy H. Sarma.
- Published
- 2024
- Full Text
- View/download PDF
16. The Polarity of an Amino Acid at Position 1891 of Severe Fever with Thrombocytopenia Syndrome Virus L Protein Is Critical for the Polymerase Activity
- Author
-
Kisho Noda, Yoshimi Tsuda, Fumiya Kozawa, Manabu Igarashi, Kenta Shimizu, Jiro Arikawa, and Kumiko Yoshimatsu
- Subjects
SFTSV ,L protein ,bunyavirus ,polymerase activity ,Microbiology ,QR1-502 - Abstract
Severe fever with thrombocytopenia syndrome virus subclone B7 shows strong plaque formation and cytopathic effect induction compared with other subclones and the parental strain YG1. Compared to YG1 and the other subclones, only B7 possesses a single substitution in the L protein at the amino acid position 1891, in which N is changed to K (N1891K). In this study, we evaluate the effects of this mutation on L protein activity via a cell-based minigenome assay. Substitutions of N with basic amino acids (K or R) enhanced polymerase activity, while substitutions with an acidic amino acid (E) decreased this activity. Mutation to other neutral amino acids showed no significant effect on activity. These results suggest that the characteristic of the amino acid at position 1891 of the L protein are critical for its function, especially with respect to the charge status. Our data indicate that this C-terminal domain of the L protein may be crucial to its functions in genome transcription and viral replication.
- Published
- 2020
- Full Text
- View/download PDF
17. Complementary Mutations in the N and L Proteins for Restoration of Viral RNA Synthesis.
- Author
-
Weike Li, Gumpper, Ryan H., Yusuf Uddin, Schmidt-Krey, Ingeborg, and Ming Luo
- Subjects
- *
RNA synthesis , *RNA viruses , *RNA replicase , *RNA polymerases , *NUCLEOCAPSIDS , *VESICULAR stomatitis , *GENETIC transcription , *VIRAL replication , *VIRUSES - Abstract
During viral RNA synthesis by the viral RNA-dependent RNA polymerase (vRdRp) of vesicular stomatitis virus, the sequestered RNA genome must be released from the nucleocapsid in order to serve as the template. Unveiling the sequestered RNA by interactions of vRdRp proteins, the large subunit (L) and the phosphoprotein (P), with the nucleocapsid protein (N) must not disrupt the nucleocapsid assembly. We noticed that a flexible structural motif composed of an α-helix and a loop in the N protein may act as the access gate to the sequestered RNA. This suggests that local conformational changes in this structural motif may be induced by interactions with the polymerase to unveil the sequestered RNA, without disrupting the nucleocapsid assembly. Mutations of several residues in this structural motif-Glu169, Phe171, and Leu174-to Ala resulted in loss of viral RNA synthesis in a minigenome assay. After implementing these mutations in the viral genome, mutant viruses were recovered by reverse genetics and serial passages. Sequencing the genomes of the mutant viruses revealed that compensatory mutations in L, P, and N were required to restore the viral viability. Corresponding mutations were introduced in L, P, and N, and their complementarity to the N mutations was confirmed by the minigenome assay. Introduction of the corresponding mutations is also sufficient to rescue the mutant viruses. These results suggested that the interplay of the N structural motif with the L protein may play a role in accessing the nucleotide template without disrupting the overall structure of the nucleocapsid. IMPORTANCE During viral RNA synthesis of a negative-strand RNA virus, the viral RNA-dependent RNA polymerase (vRdRp) must gain access to the sequestered RNA in the nucleocapsid to use it as the template, but at the same time may not disrupt the nucleocapsid assembly. Our structural and mutagenesis studies showed that a flexible structural motif acts as a potential access gate to the sequestered RNA and plays an essential role in viral RNA synthesis. Interactions of this structural motif within the vRdRp may be required for unveiling the sequestered RNA. This mechanism of action allows the sequestered RNA to be released locally without disrupting the overall structure of the nucleocapsid. Since this flexible structural motif is present in the N proteins of many NSVs, release of the sequestered RNA genome by local conformational changes in the N protein may be a general mechanism in NSV viral RNA synthesis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
18. Vesiculopolins, a New Class of Anti-Vesiculoviral Compounds, Inhibit Transcription Initiation of Vesiculoviruses
- Author
-
Minako Ogino, Yuriy Fedorov, Drew J. Adams, Kazuma Okada, Naoto Ito, Makoto Sugiyama, and Tomoaki Ogino
- Subjects
vesicular stomatitis virus ,Chandipura virus ,vesiculoviruses ,L protein ,RNA-dependent RNA polymerase ,transcription ,small molecule inhibitor ,oncolytic viruses ,vaccine vectors ,Microbiology ,QR1-502 - Abstract
Vesicular stomatitis virus (VSV) represents a promising platform for developing oncolytic viruses, as well as vaccines against significant human pathogens. To safely control VSV infection in humans, small-molecule drugs that selectively inhibit VSV infection may be needed. Here, using a cell-based high-throughput screening assay followed by an in vitro transcription assay, compounds with a 7-hydroxy-6-methyl-3,4-dihydroquinolin-2(1H)-one structure and an aromatic group at position 4 (named vesiculopolins, VPIs) were identified as VSV RNA polymerase inhibitors. The most effective compound, VPI A, inhibited VSV-induced cytopathic effects and in vitro mRNA synthesis with micromolar to submicromolar 50% inhibitory concentrations. VPI A was found to inhibit terminal de novo initiation rather than elongation for leader RNA synthesis, but not mRNA capping, with the VSV L protein, suggesting that VPI A is targeted to the polymerase domain in the L protein. VPI A inhibited transcription of Chandipura virus, but not of human parainfluenza virus 3, suggesting that it specifically acts on vesiculoviral L proteins. These results suggest that VPIs may serve not only as molecular probes to elucidate the mechanisms of transcription of vesiculoviruses, but also as lead compounds to develop antiviral drugs against vesiculoviruses and other related rhabdoviruses.
- Published
- 2019
- Full Text
- View/download PDF
19. Transcriptional Control and mRNA Capping by the GDP Polyribonucleotidyltransferase Domain of the Rabies Virus Large Protein
- Author
-
Tomoaki Ogino and Todd J. Green
- Subjects
rabies virus ,L protein ,transcription ,mRNA capping ,GDP polyribonucleotidyltransferase ,de novo initiation ,Microbiology ,QR1-502 - Abstract
Rabies virus (RABV) is a causative agent of a fatal neurological disease in humans and animals. The large (L) protein of RABV is a multifunctional RNA-dependent RNA polymerase, which is one of the most attractive targets for developing antiviral agents. A remarkable homology of the RABV L protein to a counterpart in vesicular stomatitis virus, a well-characterized rhabdovirus, suggests that it catalyzes mRNA processing reactions, such as 5′-capping, cap methylation, and 3′-polyadenylation, in addition to RNA synthesis. Recent breakthroughs in developing in vitro RNA synthesis and capping systems with a recombinant form of the RABV L protein have led to significant progress in our understanding of the molecular mechanisms of RABV RNA biogenesis. This review summarizes functions of RABV replication proteins in transcription and replication, and highlights new insights into roles of an unconventional mRNA capping enzyme, namely GDP polyribonucleotidyltransferase, domain of the RABV L protein in mRNA capping and transcription initiation.
- Published
- 2019
- Full Text
- View/download PDF
20. Transcription and replication mechanisms of Bunyaviridae and Arenaviridae L proteins.
- Author
-
Ferron, François, Weber, Friedemann, de la Torre, Juan Carlos, and Reguera, Juan
- Subjects
- *
GENETIC transcription , *RNA replicase , *ARENAVIRUSES , *NUCLEOPROTEINS , *BUNYAVIRUSES , *RNA synthesis , *RNA-protein interactions - Abstract
Bunyaviridae and Arenaviridae virus families include an important number of highly pathogenic viruses for humans. They are enveloped viruses with negative stranded RNA genomes divided into three (bunyaviruses) or two (arenaviruses) segments. Each genome segment is coated by the viral nucleoproteins (NPs) and the polymerase (L protein) to form a functional ribonucleoprotein (RNP) complex. The viral RNP provides the necessary context on which the L protein carries out the biosynthetic processes of RNA replication and gene transcription. Decades of research have provided a good understanding of the molecular processes underlying RNA synthesis, both RNA replication and gene transcription, for these two families of viruses. In this review we will provide a global view of the common features, as well as differences, of the molecular biology of Bunyaviridae and Arenaviridae . We will also describe structures of protein and protein-RNA complexes so far determined for these viral families, mainly focusing on the L protein, and discuss their implications for understanding the mechanisms of viral RNA replication and gene transcription within the architecture of viral RNPs, also taking into account the cellular context in which these processes occur. Finally, we will discuss the implications of these structural findings for the development of antiviral drugs to treat human diseases caused by members of the Bunyaviridae and Arenaviridae families. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
21. 5'-Phospho-RNA Acceptor Specificity of GDP Polyribonucleotidyltransferase of Vesicular Stomatitis Virus in mRNA Capping.
- Author
-
Minako Ogino and Tomoaki Ogino
- Subjects
- *
RHABDOVIRUSES , *GUANYLYLTRANSFERASE , *EUKARYOTIC cells , *MESSENGER RNA , *METHYLGUANOSINE - Abstract
The GDP polyribonucleotidyltransferase (PRNTase) domain of the multifunctional L protein of rhabdoviruses, such as vesicular stomatitis virus (VSV) and rabies virus, catalyzes the transfer of 5'-phospho-RNA (pRNA) from 5'-triphospho-RNA (pppRNA) to GDP via a covalent enzyme-pRNA intermediate to generate a 5'-cap structure (GpppA). Here, using an improved oligo-RNA capping assay with the VSV L protein, we showed that the Michaelis constants for GDP and pppAACAG (VSV mRNA-start sequence) are 0.03 and 0.4 µM, respectively. A competition assay between GDP and GDP analogues in the GpppA formation and pRNA transfer assay using GDP analogues as pRNA acceptors indicated that the PRNTase domain recognizes the C-2-amino group, but not the C-6-oxo group, N-1-hydrogen, or N-7-nitrogen, of GDP for the cap formation. 2,6-Diaminopurine-riboside (DAP), 7-deazaguanosine (7-deaza-G), and 7-methylguanosine (m7G) diphosphates efficiently accepted pRNA, resulting in the formation of DAPpppA, 7-deaza-GpppA, and m7GpppA (cap 0), respectively. Furthermore, either the 2'- or 3'-hydroxyl group of GDP was found to be required for efficient pRNA transfer. A 5'-diphosphate form of antiviral ribavirin weakly inhibited the GpppA formation but did not act as a pRNA acceptor. These results indicate that the PRNTase domain has a unique guanosine-binding mode different from that of eukaryotic mRNA capping enzyme, guanylyltransferase. IMPORTANCE: mRNAs of nonsegmented negative-strand (NNS) RNA viruses, such as VSV, possess a fully methylated cap structure, which is required for mRNA stability, efficient translation, and evasion of antiviral innate immunity in host cells. GDP polyribonucleotidyltransferase (PRNTase) is an unconventional mRNA capping enzyme of NNS RNA viruses that is distinct from the eukaryotic mRNA capping enzyme, guanylyltransferase. In this study, we studied the pRNA acceptor specificity of VSV PRNTase using various GDP analogues and identified chemical groups of GDP as essential for the substrate activity. The findings presented here are useful not only for understanding the mechanism of the substrate recognition with PRNTase but also for designing antiviral agents targeting this enzyme. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
22. Ebola virus: A gap in drug design and discovery - experimental and computational perspective.
- Author
-
Balmith, Marissa, Faya, Mbuso, and Soliman, Mahmoud E. S.
- Subjects
- *
EBOLA virus , *DRUG development , *EBOLA virus disease , *DRUGS , *DIAGNOSIS ,TREATMENT of Ebola virus diseases - Abstract
The Ebola virus, formally known as the Ebola hemorrhagic fever, is an acute viral syndrome causing sporadic outbreaks that have ravaged West Africa. Due to its extreme virulence and highly transmissible nature, Ebola has been classified as a category A bioweapon organism. Only recently have vaccine or drug regimens for the Ebola virus been developed, including Zmapp and peptides. In addition, existing drugs which have been repurposed toward anti-Ebola virus activity have been re-examined and are seen to be promising candidates toward combating Ebola. Drug development involving computational tools has been widely employed toward target-based drug design. Screening large libraries have greatly stimulated research toward effective anti-Ebola virus drug regimens. Current emphasis has been placed on the investigation of host proteins and druggable viral targets. There is a huge gap in the literature regarding guidelines in the discovery of Ebola virus inhibitors, which may be due to the lack of information on the Ebola drug targets, binding sites, and mechanism of action of the virus. This review focuses on Ebola virus inhibitors, drugs which could be repurposed to combat the Ebola virus, computational methods which study drug-target interactions as well as providing further insight into the mode of action of the Ebola virus. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. The Complete Sequence of a Human Parainfluenzavirus 4 Genome
- Author
-
Carmen Yea, Rose Cheung, Carol Collins, Dena Adachi, John Nishikawa, and Raymond Tellier
- Subjects
human parainfluenza virus 4 ,complete genome sequence ,L protein ,long RT-PCR ,Microbiology ,QR1-502 - Abstract
Although the human parainfluenza virus 4 (HPIV4) has been known for a long time, its genome, alone among the human paramyxoviruses, has not been completely sequenced to date. In this study we obtained the first complete genomic sequence of HPIV4 from a clinical isolate named SKPIV4 obtained at the Hospital for Sick Children in Toronto (Ontario, Canada). The coding regions for the N, P/V, M, F and HN proteins show very high identities (95% to 97%) with previously available partial sequences for HPIV4B. The sequence for the L protein and the non-coding regions represent new information. A surprising feature of the genome is its length, more than 17 kb, making it the longest genome within the genus Rubulavirus, although the length is well within the known range of 15 kb to 19 kb for the subfamily Paramyxovirinae. The availability of a complete genomic sequence will facilitate investigations on a respiratory virus that is still not completely characterized.
- Published
- 2009
- Full Text
- View/download PDF
24. Characterization of an orally available respiratory syncytial virus L protein polymerase inhibitor DZ7487.
- Author
-
Guo Q, Qian J, Zeng Q, Zhang L, Zhu X, Zheng J, Chen K, and Liu E
- Abstract
Objectives: Respiratory Syncytial Virus (RSV) is a leading cause of death and hospitalization among infants and young children. People with an immunocompromised status are also at risk for severe RSV infection. There is no specific treatment for RSV infection available. Ribavirin, an antiviral drug approved for severe lung infection by RSV, has shown limited clinical efficacies with severe side effects. Additionally, given the genetic variability of RSV genomes and seasonal change of different strains, a broad-spectrum antiviral drug is highly desirable. The RNA-dependent RNA polymerase (RdRp) domain is relatively conserved and indispensable for the replication of the virus genome and therefore serves as a potential therapeutic target. Previous attempts to identify an RdRp inhibitor have not been successful due to lack of potency or high enough blood exposure. DZ7487 is a novel orally available small molecule inhibitor specifically designed to target the RSV RdRp. Here we present our data showing that DZ7487 can potently inhibited all clinical viral isolates tested, with large safety margin predicted for human., Methods: HEp-2 cells were infected by RSV A and B. Antiviral activities were assessed by in vitro cytopathic effect assay (CPE) and Reverse transcription-quantitative polymerase chain reaction (RT-qPCR). DZ7487 antiviral effects in lower airway cells were evaluated in A549 and human small airway epithelial cells (SAEC) cells. DZ7487 induced RSV A2 escape mutations were selected through continuous culture with increasing DZ7487 concentrations in the culture medium. Resistant mutations were identified by next generation sequencing and confirmed by recombinant RSV CPE assays. RSV infection models in both BALB/c mice and cotton rats were used to evaluate DZ7487 in vivo antiviral effects., Results: DZ7487 potently inhibited viral replication of all clinical isolates of both RSVA and B subtypes. In lower airway cells, DZ7487 showed superior efficacy than the nucleoside analog ALS-8112. Acquired resistant mutation was predominantly restricted at the RdRp domain resulting asparagine to threonine mutation (N363T) of the L protein. This finding is consistent with DZ7487's presumed binding mode. DZ7487 was well tolerated in animal models. Unlike fusion inhibitors, which can only prevent viral infection, DZ7487 potently inhibited RSV replication before and after RSV infection in vitro and in vivo ., Conclusions: DZ7487 demonstrated potent anti-RSV replication effect both in vitro and in vivo assays. It has the desired drug-like physical properties to be an effective orally available anti-RSV replication drug with broad spectrum., Competing Interests: DZ7487 is an investigational drug of Dizal Pharmaceuticals., (AJTR Copyright © 2023.)
- Published
- 2023
25. Sequence analysis of the L protein of the Ebola 2014 outbreak: Insight into conserved regions and mutations.
- Author
-
AYUB, GOHAR and WAHEED, YASIR
- Subjects
- *
EBOLA virus , *EBOLA virus disease , *DISEASE outbreaks , *PHYLOGENY , *NUCLEOTIDE sequencing - Abstract
The 2014 Ebola outbreak was one of the largest that have occurred; it started in Guinea and spread to Nigeria, Liberia and Sierra Leone. Phylogenetic analysis of the current virus species indicated that this outbreak is the result of a divergent lineage of the Zaire ebolavirus. The L protein of Ebola virus (EBOV) is the catalytic subunit of the RNA-dependent RNA polymerase complex, which, with VP35, is key for the replication and transcription of viral RNA. Earlier sequence analysis demonstrated that the L protein of all non-segmented negative-sense (NNS) RNA viruses consists of six domains containing conserved functional motifs. The aim of the present study was to analyze the presence of these motifs in 2014 EBOV isolates, highlight their function and how they may contribute to the overall pathogenicity of the isolates. For this purpose, 81 2014 EBOV L protein sequences were aligned with 475 other NNS RNA viruses, including Paramyxoviridae and Rhabdoviridae viruses. Phylogenetic analysis of all EBOV outbreak L protein sequences was also performed. Analysis of the amino acid substitutions in the 2014 EBOV outbreak was conducted using sequence analysis. The alignment demonstrated the presence of previously conserved motifs in the 2014 EBOV isolates and novel residues. Notably, all the mutations identified in the 2014 EBOV isolates were tolerant, they were pathogenic with certain examples occurring within previously determined functional conserved motifs, possibly altering viral pathogenicity, replication and virulence. The phylogenetic analysis demonstrated that all sequences with the exception of the 2014 EBOV sequences were clustered together. The 2014 EBOV outbreak has acquired a great number of mutations, which may explain the reasons behind this unprecedented outbreak. Certain residues critical to the function of the polymerase remain conserved and may be targets for the development of antiviral therapeutic agents. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
26. The Pathogenesis of Saffold Virus in AG129 Mice and the Effects of Its Truncated L Protein in the Central Nervous System.
- Author
-
Shawn Zheng Kai Tan, Kaw Bing Chua, Yishi Xu, and Prabakaran, Mookkan
- Subjects
- *
CENTRAL nervous system diseases , *VIRUSES , *NERVOUS system , *ENCEPHALOMYELITIS , *BLOOD-brain barrier disorders - Abstract
Saffold Virus (SAFV) is a human cardiovirus that has been suggested to cause severe infection of the central nervous system (CNS). Compared to a similar virus, Theiler's murine encephalomyelitis virus (TMEV), SAFV has a truncated Leader (L) protein, a protein essential in the establishment of persistent CNS infections. In this study, we generated a chimeric SAFV by replacing the L protein of SAFV with that of TMEV. We then compared the replication in cell cultures and pathogenesis in a mouse model. We showed that both SAFV and chimeric SAFV are able to infect Vero and Neuro2a cells well, but only chimeric SAFV was able to infect RAW264.7. We then showed that mice lacking IFN-α/α and IFN-γ receptors provide a good animal model for SAFV infection, and further identified the locality of the infection to the ventral horn of the spine and several locations in the brain. Lastly, we showed that neither SAFV nor chimeric SAFV causes persistence in this model. Overall, our results provide a strong basis on which the mechanisms underlying Saffold virus induced neuropathogenesis can be further studied and, hence, facilitating new information about its pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
27. Evidence for N guanine methyl transferase activity encoded within the modular domain of RNA-dependent RNA polymerase L of a Morbillivirus.
- Author
-
Gopinath, M. and Shaila, M.
- Abstract
Post-transcriptional modification of viral mRNA is essential for the translation of viral proteins by cellular translation machinery. Due to the cytoplasmic replication of Paramyxoviruses, the viral-encoded RNA-dependent RNA polymerase (RdRP) is thought to possess all activities required for mRNA capping and methylation. In the present work, using partially purified recombinant RNA polymerase complex of rinderpest virus expressed in insect cells, we demonstrate the in vitro methylation of capped mRNA. Further, we show that a recombinant C-terminal fragment (1717-2183 aa) of L protein is capable of methylating capped mRNA, suggesting that the various post-transcriptional activities of the L protein are located in independently folding domains. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
28. Identification of two essential aspartates for polymerase activity in parainfluenza virus L protein by a minireplicon system expressing secretory luciferase.
- Author
-
Matsumoto, Yusuke, Ohta, Keisuke, Yumine, Natsuko, Goto, Hideo, and Nishio, Machiko
- Subjects
PARAINFLUENZA viruses ,ASPARTATES ,POLYMERASES ,VIRUS identification ,VIRAL proteins ,LUCIFERASES ,GENE expression in viruses - Abstract
ABSTRACT Gene expression of nonsegmented negative-strand RNA viruses (nsNSVs) such as parainfluenza viruses requires the RNA synthesis activity of their polymerase L protein; however, the detailed mechanism of this process is poorly understood. In this study, a parainfluenza minireplicon assay expressing secretory Gaussia luciferase (Gluc) was established to analyze large protein (L) activity. Measurement of Gluc expression in the culture medium of cells transfected with the minigenome and viral polymerase components enabled quick and concise calculation of L activity. By comparing the amino acid sequences in conserved region III (CRIII), a putative polymerase-active domain of the L protein, two strictly conserved aspartates were identified in all families of nsNSV. A series of L mutants from human parainfluenza virus type 2 and parainfluenza virus type 5 showed that these aspartates are necessary for reporter gene expression. It was also confirmed that these aspartates are important for the production of viral mRNA and antigenome cRNA, but not for a polymerase-complex formation. These findings suggest that these two aspartates are key players in the nucleotidyl transfer reaction using two metal ions. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
29. Interchange of L polymerase protein between two strains of viral hemorrhagic septicemia virus (VHSV) genotype IV alters temperature sensitivities in vitro.
- Author
-
Kim, Sung-Hyun, Yusuff, Shamila, Vakharia, Vikram N., and Evensen, Øystein
- Subjects
- *
POLYMERASES , *VIRAL hemorrhagic septicemia , *GENOTYPES , *IN vitro studies , *MICROBIAL genetics , *CELL lines , *VIRUS research - Abstract
Viral hemorrhagic septicemia virus (VHSV) has four genotypes (I–IV) and sub-lineages within genotype I and IV. Using a reverse genetics approach, we explored the importance of the L gene for growth characteristics at different temperatures following interchange of the L gene within genotype IV (IVa and IVb) strains. VHSV strains harboring heterologous L gene were recovered and we show that the L gene determines growth characteristics at different temperatures in permissive cell lines. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
30. The Polarity of an Amino Acid at Position 1891 of Severe Fever with Thrombocytopenia Syndrome Virus L Protein Is Critical for the Polymerase Activity
- Author
-
Yoshimi Tsuda, Jiro Arikawa, Kumiko Yoshimatsu, Manabu Igarashi, Fumiya Kozawa, Kisho Noda, and Kenta Shimizu
- Subjects
Models, Molecular ,Phlebovirus ,0301 basic medicine ,Severe Fever with Thrombocytopenia Syndrome ,Protein Conformation ,030106 microbiology ,Cell ,lcsh:QR1-502 ,Fluorescent Antibody Technique ,bunyavirus ,Genome ,Article ,lcsh:Microbiology ,Cell Line ,Viral Proteins ,03 medical and health sciences ,Transcription (biology) ,Virology ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Amino Acids ,Polymerase ,Cytopathic effect ,chemistry.chemical_classification ,biology ,Chemistry ,DNA-Directed RNA Polymerases ,Molecular biology ,polymerase activity ,Amino acid ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Amino Acid Substitution ,Viral replication ,biology.protein ,L protein ,SFTSV ,Severe fever with thrombocytopenia syndrome virus - Abstract
Severe fever with thrombocytopenia syndrome virus subclone B7 shows strong plaque formation and cytopathic effect induction compared with other subclones and the parental strain YG1. Compared to YG1 and the other subclones, only B7 possesses a single substitution in the L protein at the amino acid position 1891, in which N is changed to K (N1891K). In this study, we evaluate the effects of this mutation on L protein activity via a cell-based minigenome assay. Substitutions of N with basic amino acids (K or R) enhanced polymerase activity, while substitutions with an acidic amino acid (E) decreased this activity. Mutation to other neutral amino acids showed no significant effect on activity. These results suggest that the characteristic of the amino acid at position 1891 of the L protein are critical for its function, especially with respect to the charge status. Our data indicate that this C-terminal domain of the L protein may be crucial to its functions in genome transcription and viral replication.
- Published
- 2020
- Full Text
- View/download PDF
31. The Rabies Virus L Protein Catalyzes mRNA Capping with GDP Polyribonucleotidyltransferase Activity
- Author
-
Minako Ogino, Naoto Ito, Makoto Sugiyama, and Tomoaki Ogino
- Subjects
rabies virus ,L protein ,mRNA capping ,GDP polyribonucleotidyltransferase ,Microbiology ,QR1-502 - Abstract
The large (L) protein of rabies virus (RABV) plays multiple enzymatic roles in viral RNA synthesis and processing. However, none of its putative enzymatic activities have been directly demonstrated in vitro. In this study, we expressed and purified a recombinant form of the RABV L protein and verified its guanosine 5′-triphosphatase and GDP polyribonucleotidyltransferase (PRNTase) activities, which are essential for viral mRNA cap formation by the unconventional mechanism. The RABV L protein capped 5′-triphosphorylated but not 5′-diphosphorylated RABV mRNA-start sequences, 5′-AACA(C/U), with GDP to generate the 5′-terminal cap structure G(5′)ppp(5′)A. The 5′-AAC sequence in the substrate RNAs was found to be strictly essential for RNA capping with the RABV L protein. Furthermore, site-directed mutagenesis showed that some conserved amino acid residues (G1112, T1170, W1201, H1241, R1242, F1285, and Q1286) in the PRNTase motifs A to E of the RABV L protein are required for cap formation. These findings suggest that the putative PRNTase domain in the RABV L protein catalyzes the rhabdovirus-specific capping reaction involving covalent catalysis of the pRNA transfer to GDP, thus offering this domain as a target for developing anti-viral agents.
- Published
- 2016
- Full Text
- View/download PDF
32. Mapping of CD8 T Cell Epitopes in Human Respiratory Syncytial Virus L Protein.
- Author
-
Medina-Armenteros, Yordanka, Farinha-Arcieri, Luis E., Braga, Catarina J.M., Carromeu, Cassiano, Tamura, Rodrigo E., and Ventura, Armando M.
- Subjects
- *
RESPIRATORY syncytial virus , *CYTOTOXIC T cells , *T cells , *POLYMERASES , *AMINO acid sequence , *GLYCOPROTEINS , *PEPTIDES - Abstract
Objectives: Since it has been reported that in humans there is a relationship between human respiratory syncytial virus (hRSV)-specific cytotoxic T lymphocytes and symptom reduction, and that the polymerase (structural L protein) is highly conserved among different strains, this work aimed to identify the CD8 T cell epitopes H-2d restricted within the L sequence for immunization purposes. Methods: We screened the hRSV strain A2 L protein sequence using two independent algorithms, SYFPEITHI and PRED/BALB/c, to predict CD8 T cell epitopes. The selected peptides were synthesized and used to immunize BALB/c mice for the evaluation of T cell response. The production of IFN-γ from splenocytes of hRSV-infected animals stimulated by these peptides was assayed by ELISPOT. Results: Nine peptides showing the best binding scores to the BALB/c MHC-I molecules (H-2Kd, Ld and Dd) were selected. Sequence homology analysis showed that these sequences are conserved among different hRSV strains. Two of these peptides induced significant IFN-γ production by ex vivo-stimulated T cells. Conclusions: Our results indicate that the hRSV L protein contains H-2d-restricted epitopes. © 2014 S. Karger AG, Basel [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
33. Asymmetric packaging of polymerases within vesicular stomatitis virus.
- Author
-
Hodges, Jeffery, Tang, Xiaolin, Landesman, Michael B., Ruedas, John B., Ghimire, Anil, Gudheti, Manasa V., Perrault, Jacques, Jorgensen, Erik M., Gerton, Jordan M., and Saffarian, Saveez
- Subjects
- *
POLYMERASES , *VESICULAR stomatitis , *RNA virus infections , *PHOSPHOPROTEINS , *VIRION , *VIRAL proteins - Abstract
Highlights: [•] The VSV polymerases (L proteins) are localized to the blunt end of the virus. [•] The VSV phosphoproteins (P proteins) are localized to the blunt end of the virus. [•] Each VSV virion packages a variable number of P and L proteins. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
34. The L–VP35 and L–L interaction domains reside in the amino terminus of the Ebola virus L protein and are potential targets for antivirals.
- Author
-
Trunschke, Martina, Conrad, Dominik, Enterlein, Sven, Olejnik, Judith, Brauburger, Kristina, and Mühlberger, Elke
- Subjects
- *
EBOLA virus , *ANTIVIRAL agents , *VIRAL proteins , *RNA polymerases , *CATALYTIC RNA , *IMMUNOFLUORESCENCE , *PROTEIN binding , *AMINO acids - Abstract
Abstract: The Ebola virus (EBOV) RNA-dependent RNA polymerase (RdRp) complex consists of the catalytic subunit of the polymerase, L, and its cofactor VP35. Using immunofluorescence analysis and coimmunoprecipitation assays, we mapped the VP35 binding site on L. A core binding domain spanning amino acids 280–370 of L was sufficient to mediate weak interaction with VP35, while the entire N-terminus up to amino acid 380 was required for strong VP35–L binding. Interestingly, the VP35 binding site overlaps with an N-terminal L homo-oligomerization domain in a non-competitive manner. N-terminal L deletion mutants containing the VP35 binding site were able to efficiently block EBOV replication and transcription in a minigenome system suggesting the VP35 binding site on L as a potential target for the development of antivirals. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
35. Posttranslational modifications and secretion efficiency of immunogenic hepatitis B virus L protein deletion variants.
- Author
-
Niedre-Otomere, Baiba, Bogdanova, Ance, Bruvere, Ruta, Ose, Velta, Gerlich, Wolfram H., Pumpens, Paul, Glebe, Dieter, and Kozlovska, Tatjana
- Subjects
- *
HEPATITIS B , *VIRAL proteins , *DELETION mutation , *IMMUNOGENETICS , *IMMUNOGLOBULINS , *IMMUNIZATION , *LABORATORY mice - Abstract
Background: Subviral particles of hepatitis B virus (HBV) composed of L protein deletion variants with the 48 N-terminal amino acids of preS joined to the N-terminus of S protein (1-48preS/S) induced broadly neutralizing antibodies after immunization of mice with a Semliki Forest virus vector. A practical limitation for use as vaccine is the suboptimal secretion of such particles. The role of the N-terminal preS myristoylation in the cellular retention of full-length L protein is described controversially in the literature and the relation of these data to the truncated L protein was unknown. Thus, we studied the effect of preS myristoylation signal suppression on 1-48preS/S secretion efficiency, glycosylation and subcellular distribution. Findings: The findings are that 1-48preS/S is secreted, and that removal of the N-terminal myristoylation signal in its G2A variant reduced secretion slightly, but significantly. The glycosylation pattern of 1-48preS/S was not affected by the removal of the myristoylation signal (G2A mutant) but was different than natural L protein, whereby N4 of the preS and N3 of the S domain were ectopically glycosylated. This suggested cotranslational translocation of 1-48preS in contrast to natural L protein. The 1-48preS/S bearing a myristoylation signal was localized in a compact, perinuclear pattern with strong colocalization of preS and S epitopes, while the non-myristoylated mutants demonstrated a dispersed, granular cytoplasmic distribution with weaker colocalization. Conclusions: The large deletion in 1-48preS/S in presence of the myristoylation site facilitated formation and secretion of protein particles with neutralizing preS1 epitopes at their surface and could be a useful feature for future hepatitis B vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
36. A substitution in the pre-S1 promoter region is associated with the viral regulation of hepatitis B virus
- Author
-
Ogura, Suguru, Tameda, Masahiko, Sugimoto, Kazushi, Ikejiri, Makoto, Usui, Masanobu, Ito, Masaaki, and Takei, Yoshiyuki
- Published
- 2019
- Full Text
- View/download PDF
37. Molecular architecture of the vesicular stomatitis virus RNA polymerase.
- Author
-
Rahmeh, Amal A., Schenk, Andreas D., Danek, Eric I., Kranzusch, Philip J., Bo Liang, Walz, Thomas, and Whelan, Sean P. J.
- Subjects
- *
VESICULAR stomatitis , *RNA polymerases , *STOMATITIS in animals , *TRANSFERASES , *NUCLEOTIDES , *POLYMERIZATION - Abstract
Nonsegmented negative-strand (NNS) RNA viruses initiate infection by delivering into the host cell a highly specialized RNA synthesis machine comprising the genomic RNA completely encapsidated by the viral nucleocapsid protein and associated with the viral polymerase. The catalytic core of this protein-RNA complex is a 250-kDa multifunctional large (L) polymerase protein that contains enzymatic activities for nucleotide polymerization as well as for each step of mRNA cap formation, Working with vesicular stomatitis virus (VSV), a prototype of NNS RNA viruses, we used negative stain electron microscopy (EM) to obtain a molecular view of L, alone and in complex with the viral phosphoprotein (P) cofactor. EM analysis, combined with proteolytic digestion and deletion mapping, revealed the organization of L into a ring domain containing the RNA polymerase and an appendage of three globular domains con- taining the cap-forming activities. The capping enzyme maps to a globular domain, which is juxtaposed to the ring, and the cap methyltransferase maps to a more distal and flexibly connected globule. Upon P binding, L undergoes a significant rearrangement that may reflect an optimal positioning of its functional domains for transcription. The structural map of L provides new insights into the interrelationship of its various domains, and their rearrangement on P binding that is likely important for RNA synthesis. Because the arrangement of conserved regions involved in catalysis is homologous, the structural insights obtained for VSV L likely extend to all NNS RNA viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
38. Histidine-mediated RNA transfer to GDP for unique mRNA capping by vesicular stomatitis virus RNA polymerase.
- Author
-
Ogino, Tomoaki, Yadav, Satya P., and Banerjee, Amiya K.
- Subjects
- *
HISTIDINE , *RNA viruses , *RNA polymerases , *MUTAGENESIS , *ANTIVIRAL agents , *NEGATIVE-strand RNA viruses - Abstract
The RNA-dependent RNA polymerase L protein of vesicular stomatitis virus, a prototype of nonsegmented negative-strand (NNS) RNA viruses, forms a covalent complex with a 5′-phosphorylated viral mRNA-start sequence (L-pRNA), a putative intermediate in the unconventional mRNA capping reaction catalyzed by the RNA:GDP polyribonucleotidyltransferase (PRNTase) activity. Here, we directly demonstrate that the purified L-pRNA complex transfers pRNA to GDP to produce the capped RNA (Gpp-pRNA). indicating that the complex is a bona fide intermediate in the RNA transfer reaction. To locate the active site of the PRNTase domain in the L protein, the covalent RNA attachment site was mapped. We found that the 5′-monophosphate end of the RNA is linked to the histidine residue at position 1,227 (H1227) of the L protein through a phosphoamide bond. Interestingly, H1227 is part of the histidine-arginine (HR) motif, which is conserved within the I proteins of the NNS RNA viruses including rabies, measles, Ebola, and Borna disease viruses. Mutagenesis analyses revealed that the HR motif is required for the PRNTase activity at the step of the enzyme-pRNA intermediate formation. Thus, our findings suggest that an ancient NNS RNA viral polymerase has acquired the PRNTase domain independently of the eukaryotic mRNA capping enzyme during evolution and PRNTase becomes a rational target for designing antiviral agents. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
39. A temperature sensitive VSV identifies L protein residues that affect transcription but not replication
- Author
-
Galloway, Summer E. and Wertz, Gail W.
- Subjects
- *
VESICULAR stomatitis , *VIRAL replication , *GENETIC transcription regulation , *VIRAL proteins , *VIRAL genetics , *TEMPERATURE effect , *NUCLEOTIDE sequence - Abstract
Abstract: To investigate the polymerase components selectively involved in transcription versus replication of vesicular stomatitis virus (VSV), we sequenced the polymerase gene of a conditionally RNA defective, temperature sensitive VSV: ts(G)114, which has a phenotype upon shift from permissive to non-permissive temperature of shut-down of mRNA transcription and unaffected genome replication. Sequence analysis of the ts(G)114 L gene identified three altered amino acid residues in the L protein. These three changes were specifically engineered individually and in combinations into a functional cDNA clone encoding the VSV genome and tested for association with the temperature sensitive and RNA defective phenotypes in the background of recovered engineered viruses. The data presented in this study show a specific amino acid substitution in domain II of the VSV L protein that significantly affects total RNA synthesis, but when in combination with two additional amino acid substitutions identified in the ts(G)114 L protein, leads to a specific reduction in mRNA transcription, but not replication. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
40. Analysis of a structural homology model of the 2′-O-ribose methyltransferase domain within the vesicular stomatitis virus L protein
- Author
-
Galloway, Summer E., Richardson, Paul E., and Wertz, Gail W.
- Subjects
- *
METHYLTRANSFERASES , *VESICULAR stomatitis , *RIBOSE , *HOMOLOGY (Biology) , *RNA viruses , *TRANSCRIPTION factors , *MESSENGER RNA - Abstract
Abstract: The large (L) proteins of non-segmented negative stranded (NNS) RNA viruses contain the core RNA dependent RNA polymerase activity for RNA replication and transcription as well as the activities for polyadenylating and capping the mRNA transcripts and for methylating the cap structures. There is currently no structural information available for these large multi-functional proteins. Phylogenetic analyses have led to the division of the L protein primary structure into six functional domains of high conservation that are linked by variable regions. The studies in this report investigate the role of specific amino acids within domain VI of the VSV L protein, which contains a 2′-O-ribose methyltransferase (MTase) domain. We generated a structural homology model of residues 1644–1842 within domain VI based on the crystal structure determined for the known 2′-O-ribose MTase of E. coli, RrmJ. The information generated by this homology model directed us to residues structurally important for MTase activity and SAM binding. Selected residues were analyzed by site-specific mutagenesis and the mutant L proteins were assayed for their effects on RNA synthesis and cap methylation. The goal of this study was to functionally test the model in order to gain insight into the structural constraints of this region of the L protein. The data presented here revealed specific mutations that affect transcription, replication, and 5′ cap methylation, many of which resulted in polymerases temperature sensitive for RNA synthesis. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
41. Inhibition of measles virus and subacute sclerosing panencephalitis virus by RNA interference
- Author
-
Otaki, Momoko, Sada, Kiyonao, Kadoya, Hiroyasu, Nagano-Fujii, Motoko, and Hotta, Hak
- Subjects
- *
SUBACUTE sclerosing panencephalitis , *MEASLES virus , *RIBAVIRIN , *PARAMYXOVIRUSES - Abstract
Abstract: Subacute sclerosing panencephalitis (SSPE) is a rare, but fatal outcome of measles virus (MeV) infection. SSPE develops after prolonged persistence of mutated MeV called SSPE virus. Although a combination therapy using interferon and inosiplex or ribavirin appears to prolong survival time to some extent, there is currently no effective treatment to completely cure SSPE and a new treatment strategy is greatly needed. In this study, we adopted RNA interference (RNAi) strategy and examined whether small interfering RNAs (siRNAs) can be used to inhibit replication of MeV and SSPE virus. We report here that siRNAs targeted against L mRNA of MeV, either synthetic siRNAs or those generated by pcPUR+U6i-based expression plasmids, effectively and specifically inhibited replication of both MeV and SSPE virus without exhibiting any cytotoxic effect. The L protein of MeV is a major component of RNA-dependent RNA polymerase that is essential for viral RNA replication, and yet it is least abundant among all the MeV proteins expressed. Therefore, mRNA encoding the L protein would be a good target for RNAi strategy. The present results imply the possibility that our siRNAs against MeV L mRNA are among the potential candidates to be used to treat patients with SSPE. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
42. Genome sequence of the thermostable Newcastle disease virus (strain I-2) reveals a possible phenotypic locus
- Author
-
Kattenbelt, J.A., Meers, J., and Gould, A.R.
- Subjects
- *
NEWCASTLE disease virus , *VIRUS diseases in poultry , *BACTERIAL vaccines , *LIVESTOCK genetics - Abstract
Abstract: The complete genome sequence of the Australian I-2 heat-tolerant Newcastle disease virus (NDV) vaccine (master seed stocks) was determined and compared to the sequence of the parent virus from which it had been derived after exposure of the parent stock at 56°C for 30min. Nucleotide changes were observed at a number of positions with synonymous mutations being greater than those observed for non-synonymous mutations. Sequence data for the HN gene of a parental culture of V4 and two heat-tolerant variants of V4 were obtained. These were compared with the data for the I-2 viruses and with published sequences for parental V4 and for a number of ND vaccine strains. Sequence analyses did not reveal the ARG303 deletion in the HN protein, previously claimed to be responsible for the thermostable phenotype. No consistent changes were detected that would indicate involvement of the HN protein in heat resistance. The majority of alterations were observed in the L protein of the virus and it is proposed that these alterations were responsible for the heat-tolerant phenotype of the I-2 NDV vaccine. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
43. YM-53403, a unique anti-respiratory syncytial virus agent with a novel mechanism of action
- Author
-
Sudo, Kenji, Miyazaki, Yoji, Kojima, Naoko, Kobayashi, Masayuki, Suzuki, Hiroshi, Shintani, Masafumi, and Shimizu, Yasuaki
- Subjects
- *
PARAMYXOVIRUSES , *HERPESVIRUS diseases , *GENOMES , *NUCLEIC acids - Abstract
Abstract: We performed a large-scale random screening of an in-house chemical library based on the inhibition of respiratory syncytial virus (RSV)-induced cytopathic effect on HeLa (human cervical carcinoma) cells, and found a novel and specific anti-RSV agent, 6-{4-[(biphenyl-2-ylcarbonyl) amino]benzoyl}-N-cyclopropyl-5,6-dihydro-4H-thieno[3,2-d][1]benzazepine-2-carboxamide (YM-53403). YM-53403 potently inhibited the replication of RSV strains belonging to both A and B subgroups, but not influenza A virus, measles virus, or herpes simplex virus type 1. A plaque reduction assay was used to determine the 50% effective concentration (EC50) value for YM-53403. The value, 0.20μM, was about 100-fold more potent than ribavirin. The result of a time-dependent drug addition test showed that YM-53403 inhibited the life cycle of RSV at around 8h post-infection, suggesting an inhibitory effect on early transcription and/or replication of the RSV genome. Consistent with this result, two YM-53403-resistant viruses have a single point mutation (Y1631H) in the L protein which is a RNA polymerase for both the transcription and replication of the RSV genome. YM-53403 is an attractive compound for the treatment of RSV infection because of its highly potent anti-RSV activity and the new mode of action, which differs from that of currently reported antiviral agents. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
44. Recombinant Sendai viruses with L1618V mutation in their L polymerase protein establish persistent infection, but not temperature sensitivity
- Author
-
Nishio, Machiko, Nagata, Ai, Tsurudome, Masato, Ito, Morihiro, Kawano, Mitsuo, Komada, Hiroshi, and Ito, Yasuhiko
- Subjects
- *
SENDAI virus , *PARAINFLUENZA viruses , *PHENOTYPES , *AMINO acids - Abstract
The Sendai virus pi strain (SeVpi) isolated from cells persistently infected with SeV shows mainly two phenotypes: (1) temperature sensitivity and (2) an ability of establishing persistent infection (steady state). Three amino acid substitutions are found in the Lpi protein and are located at aa 1088, 1618, and 1664. Recombinant SeV(Lpi) (rSeV(Lpi)) having all these substitutions is temperature sensitive and is capable of establishing persistent infection (steady state). rSeVs carrying the fragment containing L1618V show both phenotypes. rSeV(L1618V), in which leucine at aa 1618 is replaced with valine, has the ability of establishing persistent infection, but is not a temperature-sensitive mutant, indicating that the ability of a virus to establish persistent infection can be separated from temperature sensitivity. The amino acid change at 1618(L→V) coexisting with aa 1169 threonine is required for acquirement of a temperature-sensitive phenotype. Three amino acid substitutions are also found in the Ppi protein, but rSeV(Ppi) does not show these phenotypes. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
45. The phosphoprotein (P) and L binding sites reside in the N-terminus of the L subunit of the measles virus RNA polymerase
- Author
-
Cevik, Bayram, Holmes, David E., Vrotsos, Emmanuel, Feller, Joyce A., Smallwood, Sherin, and Moyer, Sue A.
- Subjects
- *
PARAINFLUENZA viruses , *IMMUNOGLOBULIN idiotypes , *IMMUNOSPECIFICITY , *IMINO acids - Abstract
Measles virus encodes an RNA-dependent RNA polymerase composed of the L and P proteins. Recent studies have shown that the L proteins of both Sendai virus and parainfluenza virus 3 form an L–L complex [Çevik, B., Smallwood, S., Moyer, S.A., 2003. The oligomerization domain resides at the very Nterminus of the Sendai virus L RNA polymerase protein. Virology 313, 525–536.; Smallwood, S., Moyer, S.A., 2004. The L polymerase protein of parainfluenza virus 3 forms anoligomer and can interact with the heterologous Sendai virus L, P and C proteins. Virology 318, 439–450.; Smallwood, S., Cevik, B., Moyer, S.A., 2002. Intragenic complementation and oligomerization of the L subunit of the Sendai virus RNA polymerase. Virology 304, 235–245.]. Using differentially tagged L proteins, we show here that measles L also forms an oligomer and the L–L binding site resides in the N-terminal 408 amino acids overlapping the P binding site in the same region of L. To identify amino acids important for binding P and L, site-directed mutagenesis of the L-408 protein was performed. Seven of twelve mutants in L-408 were unable to form a complex with measles P while the remainder did bind at least some P. In contrast, all of the mutants retained the ability to form the L–L complex, so different amino acids are involved in the L and P binding sites on L. Four of the 408 mutations defective in P binding were inserted into the full-length measles L protein and all retained L–L complex formation, but did not bind P. Full-length L mutants that did not bind P were also inactive in viral RNA synthesis, showing a direct correlation between P–L complex formation and activity. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
46. Development of a reconstitution system for Rinderpest virus RNA synthesis in vitro
- Author
-
Raha, Tamal, Chattopadhyay, Anasuya, and Shaila, M.S.
- Subjects
- *
RNA polymerases , *PHOSPHOPROTEINS , *CATALYSIS , *GENETIC transcription - Abstract
The RNA dependent RNA polymerase of Rinderpest virus consists of two subunits—the large protein (L) and the phosphoprotein (P), where L is thought to be responsible for the catalytic activities in association with P protein which plays multiple roles in transcription and replication. The nucleocapsid protein (N) is necessary for encapsidation of genomic RNA, which is required as N–P complex. To understand the different steps of transcription and replication as well as the roles played by the three proteins, an in vitro reconstitution system for RNA synthesis is necessary which is not available for any morbillivirus. We describe here, an in vitro reconstitution system for transcription and replication of Rinderpest virus utilizing a synthetic, positive sense N-RNA minigenome template, free of endogenous viral polymerase proteins and recombinant viral proteins (
P+L andP+N ) expressed in insect cells by recombinant baculoviruses. We show that although L–P complex is sufficient to synthesize negative sense minigenome RNA, soluble N protein is necessary for encapsidation of RNA as well as synthesis of (+) sense leader RNA and (+) sense minigenome RNA. [Copyright &y& Elsevier]- Published
- 2004
- Full Text
- View/download PDF
47. Characterization of Sendai virus persistently infected L929 cells and Sendai virus pi strain: recombinant Sendai viruses having Mpi protein shows lower cytotoxicity and are incapable of establishing persistent infection
- Author
-
Nishio, Machiko, Tsurudome, Masato, Ito, Morihiro, Kawano, Mitsuo, Komada, Hiroshi, and Ito, Yasuhiko
- Subjects
- *
SENDAI virus , *IMMOBILIZED proteins , *CELL-mediated cytotoxicity - Abstract
It is commonly accepted that the temperature-sensitive phenotype of Sendai virus (SeV) persistently infected cells is caused by the M and/or HN proteins. Expression level of the L, M, HN, and V proteins is extremely low in L929 cells persistently infected with SeVpi (L929/SeVpi cells) incubated at 38°C. The HN protein quickly disappears in L929/SeVpi cells following a temperature shift up to 38°C, and pulse–chase experiments show that the Lpi, HNpi, and Mpi proteins are unstable at 38°C. Following a temperature shift either upward or downward, M protein is translocated into the nucleus and then localizes to the perinuclear region. None of virus-specific polypeptides are detected in the cells primarily infected with SeVpi and incubated at 38°C and virus proteins are not pulse-labeled at 38°C, indicating that temperature-sensitive step is at an early stage of infection. The Mpi protein is transiently located in the nucleus of the SeVpi primarily infected cells. Recombinant SeVs possessing the HNpi or/and Mpi proteins are not temperature-sensitive. The HN protein is expressed at very low levels and the F protein localizes to the perinuclear region in rSeV(Mpi)-infected cells incubated at 38°C for 18 h. rSeVs having the Mpi protein exhibit lower cytotoxicity and are incapable of establishing persistent infection. Amino acid 116 of the Mpi protein is related to the nuclear translocation and lower cytopathogenesis, whereas aa183 is involved in the interaction between M protein and viral glycoproteins. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
48. Association of L Protein and in vitro Tomato Spotted Wilt Virus RNA-Dependent RNA Polymerase Activity.
- Author
-
Chapman, Elisabeth J., Hilson, Pierre, and German, Thomas L.
- Subjects
- *
TOMATO spotted wilt virus disease , *RNA polymerases , *RNA synthesis , *VIRUS diseases of plants , *PLANT viruses - Abstract
We have previously described an in vitro assay for RNA-dependent RNA polymerase activity in virions of tomato spotted wilt virus (TSWV). Here we report antibody inhibition of virion-associated RNA synthesis in vitro with an L-protein-specific polyclonal antibody raised against the carboxy-terminus of the L protein. In contrast, RNA synthesis was not inhibited by a heterologous antiserum and was unaffected by antiserum raised against an internal portion of the L protein. Our results directly associate the TSWV L protein, the putative viral polymerase, with RNA synthesis functions in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
49. Replication and transcription of viral RNAs by recombinant L proteins of New Jersey serotype of vesicular stomatitis virus
- Author
-
Kim, Gyoung Nyoun, Choi, Woo-young, Park, Manhoon, and Kang, C. Yong
- Subjects
- *
VESICULAR stomatitis , *RNA polymerases - Abstract
The large (L) protein of vesicular stomatitis virus (VSV), catalytic subunit of RNA-dependent RNA polymerase is responsible for the transcription and replication of VSV. The L protein of the Indiana serotype of VSV (VSVInd) has previously been cloned and expressed, and used in the reverse genetics of VSVInd. However, the cDNA clones expressing functional L proteins of the VSVNJ serotype were not available. It was necessary to obtain functional clones of the New Jersey serotype of VSV (VSVNJ) in order to study homologous viral interference. Here we report the cDNA cloning, expression, and functional analyses of L proteins from both the Hazelhurst subtype and Concan subtype of VSVNJ. The analysis of the expressed L proteins for the transcription and replication of VSV demonstrate that both VSVNJ L clones express functional RNA-dependent RNA polymerase. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
50. Vesiculopolins, a New Class of Anti-Vesiculoviral Compounds, Inhibit Transcription Initiation of Vesiculoviruses
- Author
-
Naoto Ito, Minako Ogino, Yuriy Fedorov, Tomoaki Ogino, Makoto Sugiyama, Drew J. Adams, and Kazuma Okada
- Subjects
0301 basic medicine ,Transcription, Genetic ,viruses ,030106 microbiology ,lcsh:QR1-502 ,RNA-dependent RNA polymerase ,Virus Replication ,Antiviral Agents ,Article ,Vesicular stomatitis Indiana virus ,lcsh:Microbiology ,Cell Line ,Small Molecule Libraries ,Viral Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,vesiculoviruses ,Transcription (biology) ,Cricetinae ,Virology ,RNA polymerase ,Animals ,Humans ,Polymerase ,Messenger RNA ,vaccine vectors ,biology ,small molecule inhibitor ,Vesiculovirus ,biology.organism_classification ,In vitro ,High-Throughput Screening Assays ,3. Good health ,Oncolytic virus ,030104 developmental biology ,Infectious Diseases ,chemistry ,Vesicular stomatitis virus ,oncolytic viruses ,Chandipura virus ,biology.protein ,RNA, Viral ,L protein ,vesicular stomatitis virus ,transcription ,HeLa Cells - Abstract
Vesicular stomatitis virus (VSV) represents a promising platform for developing oncolytic viruses, as well as vaccines against significant human pathogens. To safely control VSV infection in humans, small-molecule drugs that selectively inhibit VSV infection may be needed. Here, using a cell-based high-throughput screening assay followed by an in vitro transcription assay, compounds with a 7-hydroxy-6-methyl-3,4-dihydroquinolin-2(1H)-one structure and an aromatic group at position 4 (named vesiculopolins, VPIs) were identified as VSV RNA polymerase inhibitors. The most effective compound, VPI A, inhibited VSV-induced cytopathic effects and in vitro mRNA synthesis with micromolar to submicromolar 50% inhibitory concentrations. VPI A was found to inhibit terminal de novo initiation rather than elongation for leader RNA synthesis, but not mRNA capping, with the VSV L protein, suggesting that VPI A is targeted to the polymerase domain in the L protein. VPI A inhibited transcription of Chandipura virus, but not of human parainfluenza virus 3, suggesting that it specifically acts on vesiculoviral L proteins. These results suggest that VPIs may serve not only as molecular probes to elucidate the mechanisms of transcription of vesiculoviruses, but also as lead compounds to develop antiviral drugs against vesiculoviruses and other related rhabdoviruses.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.