Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Semin Cell Dev Biol ; 111: 148-155, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32665176

RESUMO

Alphaviruses such as chikungunya and western equine encephalitis viruses are important human pathogens transmitted by mosquitoes that have recently caused large epidemic and epizootic outbreaks. The epidemic potential of alphaviruses is often related to enhanced mosquito transmission. Tissue barriers and antiviral responses impose bottlenecks to viral populations in mosquitoes. Substitutions in the envelope proteins and the presence of repeated sequence elements (RSEs) in the 3'UTR of epidemic viruses were proposed to be specifically associated to efficient replication in mosquito vectors. Here, we discuss the molecular mechanisms that originated RSEs, the evolutionary forces that shape the 3'UTR of alphaviruses, and the significance of RSEs for mosquito transmission. Finally, the presence of RSEs in the 3'UTR of viral genomes appears as evolutionary trait associated to mosquito adaptation and emerges as a common feature among viruses from the alphavirus and flavivirus genera.


Assuntos
Infecções por Alphavirus/transmissão , Vírus Chikungunya/genética , Vírus da Encefalite Equina do Oeste/genética , Infecções por Flavivirus/transmissão , Flavivirus/genética , Genoma Viral , Proteínas do Envelope Viral/genética , Regiões 3' não Traduzidas , Infecções por Alphavirus/virologia , Animais , Vírus Chikungunya/classificação , Vírus Chikungunya/patogenicidade , Culicidae/virologia , Vírus da Encefalite Equina do Oeste/classificação , Vírus da Encefalite Equina do Oeste/patogenicidade , Flavivirus/classificação , Flavivirus/patogenicidade , Infecções por Flavivirus/virologia , Regulação da Expressão Gênica , Humanos , Repetições de Microssatélites , Mosquitos Vetores/virologia , Filogenia , Transdução de Sinais , Proteínas do Envelope Viral/metabolismo , Replicação Viral
2.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33148794

RESUMO

Chikungunya virus (CHIKV) is a reemerging and rapidly spreading pathogen transmitted by mosquitoes. The emergence of new epidemic variants of the virus is associated with genetic evolutionary traits, including duplication of repeated RNA elements in the 3' untranslated region (UTR) that seemingly favor transmission by mosquitoes. The transmission potential of a given variant results from a complex interplay between virus populations and anatomical tissue barriers in the mosquito. Here, we used the wild-type CHIKV Caribbean strain and an engineered mutant harboring a deletion in the 3' UTR to dissect the interactions of virus variants with the anatomical barriers that impede transmission during the replication cycle of the virus in Aedes mosquitoes. Compared to the 3'-UTR mutant, we observed that the wild-type virus had a short extrinsic incubation period (EIP) after an infectious blood meal and was expectorated into mosquito saliva much more efficiently. We found that high viral titers in the midgut are not sufficient to escape the midgut escape barrier. Rather, viral replication kinetics play a crucial role in determining midgut escape and the transmission ability of CHIKV. Finally, competition tests in mosquitoes coinfected with wild-type and mutant viruses revealed that both viruses successfully colonized the midgut, but wild-type viruses effectively displaced mutant viruses during systemic infection due to their greater efficiency of escaping from the midgut into secondary tissues. Overall, our results uncover a link between CHIKV replication kinetics and the effect of bottlenecks on population diversity, as slowly replicating variants are less able to overcome the midgut escape barrier.IMPORTANCE It is well established that selective pressures in mosquito vectors impose population bottlenecks for arboviruses. Here, we used a CHIKV Caribbean lineage mutant carrying a deletion in the 3' UTR to study host-virus interactions in vivo in the epidemic mosquito vector Aedes aegypti We found that the mutant virus had a delayed replication rate in mosquitoes, which lengthened the extrinsic incubation period (EIP) and reduced fitness relative to the wild-type virus. As a result, the mutant virus displayed a reduced capacity to cross anatomical barriers during the infection cycle in mosquitoes, thus reducing the virus transmission rate. Our findings show how selective pressures act on CHIKV noncoding regions to select variants with shorter EIPs that are preferentially transmitted by the mosquito vector.


Assuntos
Aedes/virologia , Febre de Chikungunya/transmissão , Vírus Chikungunya/patogenicidade , Trato Gastrointestinal/virologia , Interações Hospedeiro-Patógeno , Mosquitos Vetores/virologia , Replicação Viral , Animais , Vírus Chikungunya/genética , Feminino , Humanos , Mutação , Carga Viral
3.
PLoS Pathog ; 15(4): e1007706, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30986247

RESUMO

The potential of RNA viruses to adapt to new environments relies on their ability to introduce changes in their genomes, which has resulted in the recent expansion of re-emergent viruses. Chikungunya virus is an important human pathogen transmitted by mosquitoes that, after 60 years of exclusive circulation in Asia and Africa, has rapidly spread in Europe and the Americas. Here, we examined the evolution of CHIKV in different hosts and uncovered host-specific requirements of the CHIKV 3'UTR. Sequence repeats are conserved at the CHIKV 3'UTR but vary in copy number among viral lineages. We found that these blocks of repeated sequences favor RNA recombination processes through copy-choice mechanism that acts concertedly with viral selection, determining the emergence of new viral variants. Functional analyses using a panel of mutant viruses indicated that opposite selective pressures in mosquito and mammalian cells impose a fitness cost during transmission that is alleviated by recombination guided by sequence repeats. Indeed, drastic changes in the frequency of viral variants with different numbers of repeats were detected during host switch. We propose that RNA recombination accelerates CHIKV adaptability, allowing the virus to overcome genetic bottlenecks within the mosquito host. These studies highlight the role of 3'UTR plasticity on CHIKV evolution, providing a new paradigm to explain the significance of sequence repetitions.


Assuntos
Regiões 3' não Traduzidas/genética , Aedes/virologia , Febre de Chikungunya/virologia , Vírus Chikungunya/patogenicidade , RNA/genética , Recombinação Genética , Replicação Viral/genética , Aedes/genética , Animais , Sequência de Bases , Células Cultivadas , Febre de Chikungunya/genética , Febre de Chikungunya/transmissão , Evolução Molecular , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , RNA Viral/genética , Sequências Repetitivas de Ácido Nucleico
4.
mBio ; 10(1)2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30622191

RESUMO

Flaviviruses include a diverse group of medically important viruses that cycle between mosquitoes and humans. During this natural process of switching hosts, each species imposes different selective forces on the viral population. Using dengue virus (DENV) as model, we found that paralogous RNA structures originating from duplications in the viral 3' untranslated region (UTR) are under different selective pressures in the two hosts. These RNA structures, known as dumbbells (DB1 and DB2), were originally proposed to be enhancers of viral replication. Analysis of viruses obtained from infected mosquitoes showed selection of mutations that mapped in DB2. Recombinant viruses carrying the identified variations confirmed that these mutations greatly increase viral replication in mosquito cells, with low or no impact in human cells. Use of viruses lacking each of the DB structures revealed opposite viral phenotypes. While deletion of DB1 reduced viral replication about 10-fold, viruses lacking DB2 displayed a great increase of fitness in mosquitoes, confirming a functional diversification of these similar RNA elements. Mechanistic analysis indicated that DB1 and DB2 differentially modulate viral genome cyclization and RNA replication. We found that a pseudoknot formed within DB2 competes with long-range RNA-RNA interactions that are necessary for minus-strand RNA synthesis. Our results support a model in which a functional diversification of duplicated RNA elements in the viral 3' UTR is driven by host-specific requirements. This study provides new ideas for understanding molecular aspects of the evolution of RNA viruses that naturally jump between different species.IMPORTANCE Flaviviruses constitute the most relevant group of arthropod-transmitted viruses, including important human pathogens such as the dengue, Zika, yellow fever, and West Nile viruses. The natural alternation of these viruses between vertebrate and invertebrate hosts shapes the viral genome population, which leads to selection of different viral variants with potential implications for epidemiological fitness and pathogenesis. However, the selective forces and mechanisms acting on the viral RNA during host adaptation are still largely unknown. Here, we found that two almost identical tandem RNA structures present at the viral 3' untranslated region are under different selective pressures in the two hosts. Mechanistic studies indicated that the two RNA elements, known as dumbbells, contain sequences that overlap essential RNA cyclization elements involved in viral RNA synthesis. The data support a model in which the duplicated RNA structures differentially evolved to accommodate distinct functions for viral replication in the two hosts.


Assuntos
Regiões 3' não Traduzidas , Vírus da Dengue/genética , Conformação de Ácido Nucleico , RNA Viral/genética , Animais , Culicidae , Vírus da Dengue/crescimento & desenvolvimento , Especificidade de Hospedeiro , Humanos , Sequências Repetitivas de Ácido Nucleico , Seleção Genética , Replicação Viral
5.
PLoS Pathog ; 13(3): e1006265, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28264033

RESUMO

The Flavivirus genus includes a large number of medically relevant pathogens that cycle between humans and arthropods. This host alternation imposes a selective pressure on the viral population. Here, we found that dengue virus, the most important viral human pathogen transmitted by insects, evolved a mechanism to differentially regulate the production of viral non-coding RNAs in mosquitos and humans, with a significant impact on viral fitness in each host. Flavivirus infections accumulate non-coding RNAs derived from the viral 3'UTRs (known as sfRNAs), relevant in viral pathogenesis and immune evasion. We found that dengue virus host adaptation leads to the accumulation of different species of sfRNAs in vertebrate and invertebrate cells. This process does not depend on differences in the host machinery; but it was found to be dependent on the selection of specific mutations in the viral 3'UTR. Dissecting the viral population and studying phenotypes of cloned variants, the molecular determinants for the switch in the sfRNA pattern during host change were mapped to a single RNA structure. Point mutations selected in mosquito cells were sufficient to change the pattern of sfRNAs, induce higher type I interferon responses and reduce viral fitness in human cells, explaining the rapid clearance of certain viral variants after host change. In addition, using epidemic and pre-epidemic Zika viruses, similar patterns of sfRNAs were observed in mosquito and human infected cells, but they were different from those observed during dengue virus infections, indicating that distinct selective pressures act on the 3'UTR of these closely related viruses. In summary, we present a novel mechanism by which dengue virus evolved an RNA structure that is under strong selective pressure in the two hosts, as regulator of non-coding RNA accumulation and viral fitness. This work provides new ideas about the impact of host adaptation on the variability and evolution of flavivirus 3'UTRs with possible implications in virulence and viral transmission.


Assuntos
Adaptação Biológica/genética , Culicidae/virologia , Vírus da Dengue/genética , Aptidão Genética/genética , RNA Viral/genética , Regiões 3' não Traduzidas/genética , Animais , Northern Blotting , Dengue/genética , Variação Genética , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Humanos , Insetos Vetores/virologia , Filogenia , Reação em Cadeia da Polimerase , Transfecção
6.
J Virol ; 90(11): 5451-61, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27009958

RESUMO

UNLABELLED: Dengue virus is currently the most important insect-borne viral human pathogen. Viral nonstructural protein 3 (NS3) is a key component of the viral replication machinery that performs multiple functions during viral replication and participates in antiviral evasion. Using dengue virus infectious clones and reporter systems to dissect each step of the viral life cycle, we examined the requirements of different domains of NS3 on viral particle assembly. A thorough site-directed mutagenesis study based on solvent-accessible surface areas of NS3 revealed that, in addition to being essential for RNA replication, different domains of dengue virus NS3 are critically required for production of infectious viral particles. Unexpectedly, point mutations in the protease, interdomain linker, or helicase domain were sufficient to abolish infectious particle formation without affecting translation, polyprotein processing, or RNA replication. In particular, we identified a novel proline-rich N-terminal unstructured region of NS3 that contains several amino acid residues involved in infectious particle formation. We also showed a new role for the interdomain linker of NS3 in virion assembly. In conclusion, we present a comprehensive genetic map of novel NS3 determinants for viral particle assembly. Importantly, our results provide evidence of a central role of NS3 in the coordination of both dengue virus RNA replication and particle formation. IMPORTANCE: Dengue virus is an important human pathogen, and its prominence is expanding globally; however, basic aspects of its biology are still unclear, hindering the development of effective therapeutic and prophylactic treatments. Little is known about the initial steps of dengue and other flavivirus particle assembly. This process involves a complex interplay between viral and cellular components, making it an attractive antiviral target. Unpredictably, we identified spatially separated regions of the large NS3 viral protein as determinants for dengue virus particle assembly. NS3 is a multifunctional enzyme that participates in different steps of the viral life cycle. Using reporter systems to dissect different viral processes, we identified a novel N-terminal unstructured region of the NS3 protein as crucial for production of viral particles. Based on our findings, we propose new ideas that include NS3 as a possible scaffold for the viral assembly process.


Assuntos
Vírus da Dengue/fisiologia , Prolina/química , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus/genética , Replicação Viral , Linhagem Celular , Vírus da Dengue/química , Vírus da Dengue/genética , Humanos , Mutagênese Sítio-Dirigida , Mutação Puntual , Prolina/metabolismo , Domínios Proteicos , RNA Helicases/química , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Viral/genética , Serina Endopeptidases/química , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Proteínas não Estruturais Virais/genética , Vírion/genética
7.
Trends Microbiol ; 24(4): 270-283, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26850219

RESUMO

Flaviviruses include a highly diverse group of arboviruses with a global distribution and a high human disease burden. Most flaviviruses cycle between insects and vertebrate hosts; thus, they are obligated to use different cellular machinery for their replication and mount different mechanisms to evade specific antiviral responses. In addition to coding for viral proteins, the viral genome contains signals in RNA structures that govern the amplification of viral components and participate in triggering or evading antiviral responses. In this review, we focused on new information about host-specific functions of RNA structures present in the 3' untranslated region (3' UTR) of flavivirus genomes. Models and conservation patterns of RNA elements of distinct flavivirus ecological groups are revised. An intriguing feature of the 3' UTR of insect-borne flavivirus genomes is the conservation of complex RNA structure duplications. Here, we discuss new hypotheses of how these RNA elements specialize for replication in vertebrate and invertebrate hosts, and present new ideas associating the significance of RNA structure duplication, small subgenomic flavivirus RNA formation, and host adaptation.


Assuntos
Flavivirus/genética , Interações Hospedeiro-Patógeno/genética , Conformação de Ácido Nucleico , RNA Viral/genética , Adaptação Biológica/genética , Animais , Evolução Molecular , Flavivirus/química , Flavivirus/metabolismo , Genoma Viral , Humanos , Filogenia , RNA Viral/biossíntese , RNA Viral/química
8.
PLoS Pathog ; 11(1): e1004604, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25635835

RESUMO

Many viral pathogens cycle between humans and insects. These viruses must have evolved strategies for rapid adaptation to different host environments. However, the mechanistic basis for the adaptation process remains poorly understood. To study the mosquito-human adaptation cycle, we examined changes in RNA structures of the dengue virus genome during host adaptation. Deep sequencing and RNA structure analysis, together with fitness evaluation, revealed a process of host specialization of RNA elements of the viral 3'UTR. Adaptation to mosquito or mammalian cells involved selection of different viral populations harvesting mutations in a single stem-loop structure. The host specialization of the identified RNA structure resulted in a significant viral fitness cost in the non-specialized host, posing a constraint during host switching. Sequence conservation analysis indicated that the identified host adaptable stem loop structure is duplicated in dengue and other mosquito-borne viruses. Interestingly, functional studies using recombinant viruses with single or double stem loops revealed that duplication of the RNA structure allows the virus to accommodate mutations beneficial in one host and deleterious in the other. Our findings reveal new concepts in adaptation of RNA viruses, in which host specialization of RNA structures results in high fitness in the adapted host, while RNA duplication confers robustness during host switching.


Assuntos
Vírus da Dengue/genética , Interações Hospedeiro-Patógeno/genética , Conformação de Ácido Nucleico , RNA Viral/química , Regiões 3' não Traduzidas , Adaptação Biológica/genética , Animais , Células Cultivadas , Cricetinae , Culicidae , Especificidade de Hospedeiro/genética , Humanos , Mutação , RNA Viral/genética
9.
J Virol ; 89(6): 3430-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25589642

RESUMO

The dengue virus genome is a dynamic molecule that adopts different conformations in the infected cell. Here, using RNA folding predictions, chemical probing analysis, RNA binding assays, and functional studies, we identified new cis-acting elements present in the capsid coding sequence that facilitate cyclization of the viral RNA by hybridization with a sequence involved in a local dumbbell structure at the viral 3' untranslated region (UTR). The identified interaction differentially enhances viral replication in mosquito and mammalian cells.


Assuntos
Proteínas do Capsídeo/genética , Vírus da Dengue/genética , Regulação Viral da Expressão Gênica , Genoma Viral , RNA Viral/química , RNA Viral/genética , Elementos Reguladores de Transcrição , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Culicidae/virologia , Replicação do DNA , Vírus da Dengue/química , Vírus da Dengue/metabolismo , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/metabolismo
10.
Viruses ; 3(9): 1739-56, 2011 09.
Artigo em Inglês | MEDLINE | ID: mdl-21994804

RESUMO

Dengue virus (DENV) genome amplification is a process that involves the viral RNA, cellular and viral proteins, and a complex architecture of cellular membranes. The viral RNA is not a passive template during this process; it plays an active role providing RNA signals that act as promoters, enhancers and/or silencers of the replication process. RNA elements that modulate RNA replication were found at the 5' and 3' UTRs and within the viral coding sequence. The promoter for DENV RNA synthesis is a large stem loop structure located at the 5' end of the genome. This structure specifically interacts with the viral polymerase NS5 and promotes RNA synthesis at the 3' end of a circularized genome. The circular conformation of the viral genome is mediated by long range RNA-RNA interactions that span thousands of nucleotides. Recent studies have provided new information about the requirement of alternative, mutually exclusive, structures in the viral RNA, highlighting the idea that the viral genome is flexible and exists in different conformations. In this article, we describe elements in the promoter SLA and other RNA signals involved in NS5 polymerase binding and activity, and provide new ideas of how dynamic secondary and tertiary structures of the viral RNA participate in the viral life cycle.


Assuntos
Vírus da Dengue/genética , Dengue/virologia , Genoma Viral/genética , RNA Viral/genética , Proteínas não Estruturais Virais/metabolismo , Vírus da Dengue/fisiologia , Humanos , Conformação de Ácido Nucleico , Regiões Promotoras Genéticas , RNA Viral/metabolismo , Regiões não Traduzidas/genética , Proteínas não Estruturais Virais/genética , Replicação Viral
11.
J Virol ; 85(12): 5745-56, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21471248

RESUMO

The mechanism by which viral RNA-dependent RNA polymerases (RdRp) specifically amplify viral genomes is still unclear. In the case of flaviviruses, a model has been proposed that involves the recognition of an RNA element present at the viral 5' untranslated region, stem-loop A (SLA), that serves as a promoter for NS5 polymerase binding and activity. Here, we investigated requirements for specific promoter-dependent RNA synthesis of the dengue virus NS5 protein. Using mutated purified NS5 recombinant proteins and infectious viral RNAs, we analyzed the requirement of specific amino acids of the RdRp domain on polymerase activity and viral replication. A battery of 19 mutants was designed and analyzed. By measuring polymerase activity using nonspecific poly(rC) templates or specific viral RNA molecules, we identified four mutants with impaired polymerase activity. Viral full-length RNAs carrying these mutations were found to be unable to replicate in cell culture. Interestingly, one recombinant NS5 protein carrying the mutations K456A and K457A located in the F1 motif lacked RNA synthesis dependent on the SLA promoter but displayed high activity using a poly(rC) template. Promoter RNA binding of this NS5 mutant was unaffected while de novo RNA synthesis was abolished. Furthermore, the mutant maintained RNA elongation activity, indicating a role of the F1 region in promoter-dependent initiation. In addition, four NS5 mutants were selected to have polymerase activity in the recombinant protein but delayed or impaired virus replication when introduced into an infectious clone, suggesting a role of these amino acids in other functions of NS5. This work provides new molecular insights on the specific RNA synthesis activity of the dengue virus NS5 polymerase.


Assuntos
Motivos de Aminoácidos/genética , Vírus da Dengue/enzimologia , Regiões Promotoras Genéticas/genética , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/química , Animais , Linhagem Celular , Cricetinae , Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Modelos Moleculares , Mutação , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
12.
J Biol Chem ; 286(9): 6929-39, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21183683

RESUMO

Dengue virus RNA-dependent RNA polymerase specifically binds to the viral genome by interacting with a promoter element known as stem-loop A (SLA). Although a great deal has been learned in recent years about the function of this promoter in dengue virus-infected cells, the molecular details that explain how the SLA interacts with the polymerase to promote viral RNA synthesis remain poorly understood. Using RNA binding and polymerase activity assays, we defined two elements of the SLA that are involved in polymerase interaction and RNA synthesis. Mutations at the top of the SLA resulted in RNAs that retained the ability to bind the polymerase but impaired promoter-dependent RNA synthesis. These results indicate that protein binding to the SLA is not sufficient to induce polymerase activity and that specific nucleotides of the SLA are necessary to render an active polymerase-promoter complex for RNA synthesis. We also report that protein binding to the viral RNA induces conformational changes downstream of the promoter element. Furthermore, we found that structured RNA elements at the 3' end of the template repress dengue virus polymerase activity in the context of a fully active SLA promoter. Using assays to evaluate initiation of RNA synthesis at the viral 3'-UTR, we found that the RNA-RNA interaction mediated by 5'-3'-hybridization was able to release the silencing effect of the 3'-stem-loop structure. We propose that the long range RNA-RNA interactions in the viral genome play multiple roles during RNA synthesis. Together, we provide new molecular details about the promoter-dependent dengue virus RNA polymerase activity.


Assuntos
RNA Polimerases Dirigidas por DNA , Vírus da Dengue/genética , Regiões Promotoras Genéticas/genética , RNA Viral , Proteínas não Estruturais Virais , Proteínas Virais , RNA Polimerases Dirigidas por DNA/química , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Metiltransferases/química , Metiltransferases/genética , Metiltransferases/metabolismo , Mutagênese/fisiologia , Conformação de Ácido Nucleico , Hibridização de Ácido Nucleico , Estrutura Terciária de Proteína , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
13.
J Virol ; 83(2): 993-1008, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19004935

RESUMO

The 5' untranslated region (5'UTR) of the dengue virus (DENV) genome contains two defined elements essential for viral replication. At the 5' end, a large stem-loop (SLA) structure functions as the promoter for viral polymerase activity. Next to the SLA, there is a short stem-loop that contains a cyclization sequence known as the 5' upstream AUG region (5'UAR). Here, we analyzed the secondary structure of the SLA in solution and the structural requirements of this element for viral replication. Using infectious DENV clones, viral replicons, and in vitro polymerase assays, we defined two helical regions, a side stem-loop, a top loop, and a U bulge within SLA as crucial elements for viral replication. The determinants for SLA-polymerase recognition were found to be common in different DENV serotypes. In addition, structural elements within the SLA required for DENV RNA replication were also conserved among different mosquito- and tick-borne flavivirus genomes, suggesting possible common strategies for polymerase-promoter recognition in flaviviruses. Furthermore, a conserved oligo(U) track present downstream of the SLA was found to modulate RNA synthesis in transfected cells. In vitro polymerase assays indicated that a sequence of at least 10 residues following the SLA, upstream of the 5'UAR, was necessary for efficient RNA synthesis using the viral 3'UTR as template.


Assuntos
Vírus da Dengue/fisiologia , Regiões Promotoras Genéticas , RNA Viral/biossíntese , RNA Viral/química , Replicação Viral , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Linhagem Celular , Cricetinae , Vírus da Dengue/genética , Modelos Moleculares , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/genética
14.
Virology ; 375(1): 223-35, 2008 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-18289628

RESUMO

Flavivirus RNA replication involves cyclization of the viral genome. A model for this process includes a promoter element at the 5' end of the genome and long-range RNA-RNA interactions. Two pairs of complementary sequences present at the ends of the viral RNA, known as 5'-3'CS and 5'-3'UAR, have been proposed to be involved in dengue virus genome cyclization. The requirement of 5'-3'CS complementarity for viral replication has been experimentally demonstrated for dengue and other mosquito borne flaviviruses. Here, we performed a functional analysis to study the role of 5'-3'UAR sequences using genomic and subgenomic dengue virus RNAs. We found that single mutations disrupting 5'-3' complementarity greatly compromised viral RNA synthesis. Although in most of the cases incorporation of compensatory mutations re-established viral RNA replication, certain nucleotides were found to be involved in alternative secondary structures also important for viral replication. In addition, mutations within 5' or 3'UAR in the context of an infectious dengue virus RNA resulted in spontaneous mutations that restored UAR base pairings. Together, we propose that specific UAR nucleotides as well as 5'-3'UAR complementarity constitute cis-acting signals involved in amplification of the dengue virus genome.


Assuntos
Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Vírus da Dengue/genética , RNA Viral/genética , RNA/genética , Replicação Viral , Animais , Linhagem Celular , Cricetinae , Vírus da Dengue/fisiologia , Modelos Moleculares , Mutação , Conformação de Ácido Nucleico , RNA Circular , RNA Complementar/genética , Supressão Genética
15.
Genes Dev ; 20(16): 2238-49, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16882970

RESUMO

The mechanisms of RNA replication of plus-strand RNA viruses are still unclear. Here, we identified the first promoter element for RNA synthesis described in a flavivirus. Using dengue virus as a model, we found that the viral RdRp discriminates the viral RNA by specific recognition of a 5' element named SLA. We demonstrated that RNA-RNA interactions between 5' and 3' end sequences of the viral genome enhance dengue virus RNA synthesis only in the presence of an intact SLA. We propose a novel mechanism for minus-strand RNA synthesis in which the viral polymerase binds SLA at the 5' end of the genome and reaches the site of initiation at the 3' end via long-range RNA-RNA interactions. These findings provide an explanation for the strict requirement of dengue virus genome cyclization during viral replication.


Assuntos
Vírus da Dengue/genética , Genoma Viral , RNA Viral/biossíntese , Sequências Reguladoras de Ácido Ribonucleico/fisiologia , Replicação Viral/fisiologia , Vírus da Dengue/fisiologia , Microscopia de Força Atômica , Conformação de Ácido Nucleico , RNA Viral/química , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/metabolismo
16.
Novartis Found Symp ; 277: 120-32; discussion 132-5, 251-3, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17319158

RESUMO

Sequences and structures present at the 5' and 3' UTRs of RNA viruses play crucial roles in the initiation and regulation of translation, RNA synthesis and viral assembly. In dengue virus, as well as in other mosquito-borne flaviviruses, the presence of complementary sequences at the ends of the genome mediate long-range RNA-RNA interactions. Dengue virus RNA displays two pairs of complementary sequences (CS and UAR) required for genome circularization and viral viability. In order to study the molecular mechanism by which these RNA-RNA interactions participate in the viral life cycle, we developed a dengue virus replicon system. RNA transfection of the replicon in mosquito and mammalian cells allows discrimination between RNA elements involved in translation and RNA synthesis. We found that mutations within CS or UAR at the 5' or 3' ends of the RNA that interfere with base pairing did not significantly affect translation of the input RNA but seriously compromised or abolished RNA synthesis. Furthermore, a systematic mutational analysis of UAR sequences indicated that, beside the role in RNA cyclization, specific nucleotides within UAR are also important for efficient RNA synthesis.


Assuntos
Vírus da Dengue/genética , RNA Viral/química , RNA Viral/genética , Replicação Viral , Animais , Pareamento de Bases , Genoma Viral , Humanos , Biossíntese de Proteínas
17.
J Biol Chem ; 278(25): 22265-71, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12660230

RESUMO

The activation of purified and phospholipid-depleted plasma membrane Ca2+-ATPase by phospholipids and ATP was studied. Enzyme activity increased with [ATP] along biphasic curves representing the sum of two Michaelis-Menten equations. Acidic phospholipids (phosphatidylinositol (PI) and phosphatidylserine (PS)) increased Vmax without affecting apparent affinities of the ATP sites. In the presence of 20 microm ATP, phosphorylation of the enzyme preincubated with Ca2+ (CaE1) was very fast (kapp congruent with 400 s-1). vo of phosphorylation of CaE1 increased with [ATP] along a Michaelis-Menten curve (Km of 15 microm) and was phospholipid-independent. Without Ca2+ preincubation (E1 + E2), vo of phosphorylation was also phospholipid-independent, but was slower and increased with [ATP] along biphasic curves. The high affinity component reflected rapid phosphorylation of CaE1, the low affinity component the E2 --> E1 shift, which accelerated to a rate higher than that of the ATPase activity when ATP was bound to the regulatory site. Dephosphorylation of EP did not occur without ATP. Dephosphorylation increased along a biphasic curve with increasing [ATP], showing that ATP accelerated dephosphorylation independently of phospholipid. PI, but not phosphatidylethanolamine (PE), accelerated dephosphorylation even in the absence of ATP. kapp for dephosphorylation was 57 s-1 at 0 microM ATP; that rate was further increased by ATP. Steady-state [EP] x kapp for dephosphorylation varied with [ATP], and matched the Ca2+-ATPase activity measured under the same conditions. Apparently, the catalytic cycle is rate-limited by dephosphorylation. Acidic phospholipids stimulate Ca2+-ATPase activity by accelerating dephosphorylation, while ATP accelerates both dephosphorylation and the conformational change from E2 to E1, further stimulating the ATPase activity.


Assuntos
Trifosfato de Adenosina/metabolismo , ATPases Transportadoras de Cálcio/sangue , Membrana Eritrocítica/enzimologia , Fosfolipídeos/farmacologia , Animais , Ativação Enzimática , Concentração de Íons de Hidrogênio , Cinética , Relação Estrutura-Atividade , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...