Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 46(22): 11952-11967, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30418631

RESUMO

RNA structures can interact with the ribosome to alter translational reading frame maintenance and promote recoding that result in alternative protein products. Here, we show that the internal ribosome entry site (IRES) from the dicistrovirus Cricket paralysis virus drives translation of the 0-frame viral polyprotein and an overlapping +1 open reading frame, called ORFx, via a novel mechanism whereby a subset of ribosomes recruited to the IRES bypasses 37 nucleotides downstream to resume translation at the +1-frame 13th non-AUG codon. A mutant of CrPV containing a stop codon in the +1 frame ORFx sequence, yet synonymous in the 0-frame, is attenuated compared to wild-type virus in a Drosophila infection model, indicating the importance of +1 ORFx expression in promoting viral pathogenesis. This work demonstrates a novel programmed IRES-mediated recoding strategy to increase viral coding capacity and impact virus infection, highlighting the diversity of RNA-driven translation initiation mechanisms in eukaryotes.


Assuntos
Dicistroviridae/genética , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Sítios Internos de Entrada Ribossomal , Biossíntese de Proteínas , Proteínas Virais/genética , Animais , Pareamento de Bases , Sequência de Bases , Linhagem Celular , Dicistroviridae/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/virologia , Conformação de Ácido Nucleico , Fases de Leitura Aberta , Poliproteínas/genética , Poliproteínas/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo
2.
J Virol ; 89(11): 5919-34, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25810541

RESUMO

UNLABELLED: Dicistroviridae are a family of RNA viruses that possesses a single-stranded positive-sense RNA genome containing two distinct open reading frames (ORFs), each preceded by an internal ribosome entry site that drives translation of the viral structural and nonstructural proteins, respectively. The type species, Cricket paralysis virus (CrPV), has served as a model for studying host-virus interactions; however, investigations into the molecular mechanisms of CrPV and other dicistroviruses have been limited as an established infectious clone was elusive. Here, we report the construction of an infectious molecular clone of CrPV. Transfection of in vitro-transcribed RNA from the CrPV clone into Drosophila Schneider line 2 (S2) cells resulted in cytopathic effects, viral RNA accumulation, detection of negative-sense viral RNA, and expression of viral proteins. Transmission electron microscopy, viral titers, and immunofluorescence-coupled transwell assays demonstrated that infectious viral particles are released from transfected cells. In contrast, mutant clones containing stop codons in either ORF decreased virus infectivity. Injection of adult Drosophila flies with virus derived from CrPV clones but not UV-inactivated clones resulted in mortality. Molecular analysis of the CrPV clone revealed a 196-nucleotide duplication within its 5' untranslated region (UTR) that stimulated translation of reporter constructs. In cells infected with the CrPV clone, the duplication inhibited viral infectivity yet did not affect viral translation or RNA accumulation, suggesting an effect on viral packaging or entry. The generation of the CrPV infectious clone provides a powerful tool for investigating the viral life cycle and pathogenesis of dicistroviruses and may further understanding of fundamental host-virus interactions in insect cells. IMPORTANCE: Dicistroviridae, which are RNA viruses that infect arthropods, have served as a model to gain insights into fundamental host-virus interactions in insect cells. Further insights into the viral molecular mechanisms are hampered due to a lack of an established infectious clone. We report the construction of the first infectious clone of the dicistrovirus, cricket paralysis virus (CrPV). We show that transfection of the CrPV clone RNA into Drosophila cells led to production of infectious particles that resemble natural CrPV virions and result in cytopathic effects and expression of CrPV proteins and RNA in infected cells. The CrPV clone should provide insights into the dicistrovirus life cycle and host-virus interactions in insect cells. Using this clone, we find that a 196-nucleotide duplication within the 5' untranslated region of the CrPV clone increased viral translation in reporter constructs but decreased virus infectivity, thus revealing a balance that interplays between viral translation and replication.


Assuntos
Regiões 5' não Traduzidas , Dicistroviridae/genética , RNA Viral/genética , Animais , Linhagem Celular , Clonagem Molecular , Efeito Citopatogênico Viral , Dicistroviridae/fisiologia , Drosophila , Microscopia Eletrônica de Transmissão , Biossíntese de Proteínas , RNA Viral/fisiologia , Análise de Sobrevida , Transcrição Gênica , Transfecção , Carga Viral , Vírion/ultraestrutura , Replicação Viral
3.
PLoS One ; 9(8): e103601, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25089704

RESUMO

Internal ribosome entry sites (IRES) are utilized by a subset of cellular and viral mRNAs to initiate translation during cellular stress and virus infection when canonical cap-dependent translation is compromised. The intergenic region (IGR) IRES of the Dicistroviridae uses a streamlined mechanism in which it can directly recruit the ribosome in the absence of initiation factors and initiates translation using a non-AUG codon. A subset of IGR IRESs including that from the honey bee viruses can also direct translation of an overlapping +1 frame gene. In this study, we systematically examined cellular conditions that lead to IGR IRES-mediated 0 and +1 frame translation in Drosophila S2 cells. Towards this, a novel bicistronic reporter that exploits the 2A "stop-go" peptide was developed to allow the detection of IRES-mediated translation in vivo. Both 0 and +1 frame translation by the IGR IRES are stimulated under a number of cellular stresses and in S2 cells infected by cricket paralysis virus, demonstrating a switch from cap-dependent to IRES-dependent translation. The regulation of the IGR IRES mechanism ensures that both 0 frame viral structural proteins and +1 frame ORFx protein are optimally expressed during virus infection.


Assuntos
DNA Intergênico/genética , Dicistroviridae/fisiologia , Biossíntese de Proteínas , Capuzes de RNA/metabolismo , Fases de Leitura/genética , Estresse Fisiológico/genética , Viroses/genética , Animais , Sequência de Bases , Linhagem Celular , Drosophila/citologia , Genes/genética , Genes Reporter , Humanos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Ribossomos/metabolismo , Viroses/virologia
4.
Nucleic Acids Res ; 42(14): 9366-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25038250

RESUMO

The dicistrovirus intergenic internal ribosome entry site (IGR IRES) directly recruits the ribosome and initiates translation using a non-AUG codon. A subset of IGR IRESs initiates translation in either of two overlapping open reading frames (ORFs), resulting in expression of the 0 frame viral structural polyprotein and an overlapping +1 frame ORFx. A U-G base pair adjacent to the anticodon-like pseudoknot of the IRES directs +1 frame translation. Here, we show that the U-G base pair is not absolutely required for +1 frame translation. Extensive mutagenesis demonstrates that 0 and +1 frame translation can be uncoupled. Ribonucleic acid (RNA) structural probing analyses reveal that the mutant IRESs adopt distinct conformations. Toeprinting analysis suggests that the reading frame is selected at a step downstream of ribosome assembly. We propose a model whereby the IRES adopts conformations to occlude the 0 frame aminoacyl-tRNA thereby allowing delivery of the +1 frame aminoacyl-tRNA to the A site to initiate translation of ORFx. This study provides a new paradigm for programmed recoding mechanisms that increase the coding capacity of a viral genome.


Assuntos
Fases de Leitura Aberta , Iniciação Traducional da Cadeia Peptídica , RNA Viral/química , Pareamento de Bases , Dicistroviridae/genética , Células HeLa , Humanos , Mutação , Conformação de Ácido Nucleico , Ribossomos/metabolismo
5.
Methods ; 59(2): 167-79, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23009811

RESUMO

Internal ribosome entry sites are RNA elements that mediate translation in a cap-independent manner. A subset of positive strand RNA viruses utilize an IRES mechanism as a viral strategy to ensure efficient viral protein synthesis. IRES elements vary in sequence, structure, and factor requirements between virus families. Here, we describe methods to determine IRES activity and approaches to study the regulation and function of IRES-mediated translation both in vitro and in vivo. Finally, we describe a new IRES-directed reporter system which exploits the 2A 'self-cleavage' or 'stop-go' peptide for optimal detection of IRES activity.


Assuntos
Técnicas Genéticas , Biossíntese de Proteínas/genética , Vírus de RNA/genética , Ribossomos/genética , Ribossomos/virologia , Animais , Sequência de Bases , Humanos , Dados de Sequência Molecular
6.
Proc Natl Acad Sci U S A ; 109(11): E630-9, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22247292

RESUMO

The dicistrovirus intergenic region internal ribosome entry site (IRES) utilizes a unique mechanism, involving P-site tRNA mimicry, to directly assemble 80S ribosomes and initiate translation at a specific non-AUG codon in the ribosomal A site. A subgroup of dicistrovirus genomes contains an additional stem-loop 5'-adjacent to the IRES and a short open reading frame (ORFx) that overlaps the viral structural polyprotein ORF (ORF2) in the +1 reading frame. Using mass spectrometry and extensive mutagenesis, we show that, besides directing ORF2 translation, the Israeli acute paralysis dicistrovirus IRES also directs ORFx translation. The latter is mediated by a UG base pair adjacent to the P-site tRNA-mimicking domain. An ORFx peptide was detected in virus-infected honey bees by multiple reaction monitoring mass spectrometry. Finally, the 5' stem-loop increases IRES activity and may couple translation of the two major ORFs of the virus. This study reveals a novel viral strategy in which a tRNA-like IRES directs precise, initiator Met-tRNA-independent translation of two overlapping ORFs.


Assuntos
Conformação de Ácido Nucleico , RNA de Transferência/química , RNA de Transferência/genética , Fases de Leitura/genética , Ribossomos/genética , Seleção Genética , Animais , Pareamento de Bases/genética , Sequência de Bases , Abelhas/virologia , DNA Complementar/genética , DNA Intergênico/genética , Dicistroviridae/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 4F em Eucariotos/metabolismo , Dados de Sequência Molecular , Iniciação Traducional da Cadeia Peptídica/genética , Pupa/virologia
7.
RNA ; 17(3): 469-77, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21224380

RESUMO

The "RNA world" hypothesis rests on the assumption that RNA polymerase ribozymes can replicate RNA without the use of protein. In the laboratory, in vitro selection has been used to create primitive versions of such polymerases. The best variant to date is a ribozyme called B6.61 that can extend a RNA primer template by 20 nucleotides (nt). This polymerase has two domains: the recently crystallized Class I ligase core, responsible for phosphodiester bond formation, and the poorly characterized accessory domain that makes polymerization possible. Here we find that the accessory domain is specified by a 37-nt bulged stem-loop structure. The accessory domain is positioned by a tertiary interaction between the terminal AL4 loop of the accessory and the J3/4 triloop found within the ligase core. This docking interaction is associated with an unwinding of the A3 and A4 helixes that appear to facilitate the correct positioning of an essential 8-nt purine bulge found between the two helices. This, together with other constraints inferred from tethering the accessory domain to a range of sites on the ligase core, indicates that the accessory domain is draped over the vertex of the ligase core tripod structure. This geometry suggests how the purine bulge in the polymerase replaces the P2 helix in the Class I ligase with a new structure that may facilitate the stabilization of incoming nucleotide triphosphates.


Assuntos
RNA Polimerases Dirigidas por DNA/química , RNA Polimerases Dirigidas por DNA/metabolismo , RNA Catalítico/química , RNA Catalítico/metabolismo , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Catálise , Ligases/metabolismo , Dados de Sequência Molecular , Conformação de Ácido Nucleico
8.
Virol J ; 6: 193, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19895695

RESUMO

The family Dicistroviridae (order Picornavirales) includes species that infect insects and other arthropods. These viruses have a linear positive-sense ssRNA genome of approximately 8-10 kb, which contains two long ORFs. The 5' ORF encodes the nonstructural polyprotein while the 3' ORF encodes the structural polyprotein. The dicistroviruses are noteworthy for the intergenic Internal Ribosome Entry Site (IGR-IRES) that mediates efficient translation initation on the 3' ORF without the requirement for initiator Met-tRNA. Acute bee paralysis virus, Israel acute paralysis virus of bees and Kashmir bee virus form a distinct subgroup within the Dicistroviridae family. In this brief report, we describe the bioinformatic discovery of a new, apparently coding, ORF in these viruses. The ORF overlaps the 5' end of the structural polyprotein coding sequence in the +1 reading frame. We also identify a potential 14-18 bp RNA stem-loop structure 5'-adjacent to the IGR-IRES. We discuss potential translation initiation mechanisms for the novel ORF in the context of the IGR-IRES and 5'-adjacent stem-loop.


Assuntos
Dicistroviridae/genética , Homologia de Genes , RNA Viral/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Abelhas/virologia , Sítios de Ligação , Israel , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Biossíntese de Proteínas , Ribossomos/metabolismo , Alinhamento de Sequência , Análise de Sequência de DNA
9.
RNA ; 11(4): 404-11, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15703441

RESUMO

Isolating the core functional elements of an RNA is normally performed during the characterization of a new RNA in order to simplify further biochemical analysis. The removal of extraneous sequence is challenging and can lead to biases that result from the incomplete sampling of deletion variants. An impartial solution to this problem is to construct a library containing a large number of deletion constructs and to select functional RNA isolates that are at least as efficient as their full-length progenitors. Here, we use nonhomologous recombination and selection to isolate the catalytic core of a pyrimidine nucleotide synthase ribozyme. A variable-length pool of approximately 10(8) recombinant molecules that included deletions, inversions, and translocations of a 271-nucleotide-long ribozyme isolate was constructed by digesting and randomly religating its DNA genome. In vitro selection for functional ribozymes was then performed in a size-dependent and a size-independent manner. The final pools had nearly equivalent catalytic rates even though their length distributions were completely different, indicating that a diverse range of deletion constructs were functionally active. Four short sequence islands, requiring as little as 81 nt of sequence, were found within all of the truncated ribozymes and could be folded into a secondary structure consisting of three helix-loops. Our findings suggest that nonhomologous recombination is a highly efficient way to isolate a ribozyme's core motif and could prove to be a useful method for evolving new ribozyme functions from pre-existing sequences in a manner that may have played an important role early in evolution.


Assuntos
Conformação de Ácido Nucleico , Mapeamento de Nucleotídeos , RNA Catalítico/química , Recombinação Genética , Tiouracila/análogos & derivados , Sequência de Bases , DNA Ligases , DNA Polimerase Dirigida por DNA , Desoxirribonuclease I , Biblioteca Gênica , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases , Nucleotídeos de Pirimidina , RNA Catalítico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Análise de Sequência de DNA , Deleção de Sequência
10.
Biotechniques ; 33(6): 1256-60, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12503310

RESUMO

Here we report the construction of a histidine-tagged T4 RNA ligase expression plasmid (pRHT4). The construct, when overexpressed in BL21 (DE3) cells, allows the preparation of large quantities of T4 RNA ligase in high purity using only a single purification column. The histidine affinity tag does not inhibit enzyme function, and we were able to purify 1-3 mg pure protein/g cell pellet. A simple purification procedure ensures that the enzyme is de-adenylated to levels comparable to those found for many commercial preparations. The purified protein has very low levels of RNase contamination and functioned normally in a variety of activity assays.


Assuntos
Cromatografia de Afinidade/métodos , Escherichia coli/química , Histidina/análise , RNA Ligase (ATP)/isolamento & purificação , Proteínas Virais/isolamento & purificação , Bacteriófago T4/genética , Sequência de Bases , Escherichia coli/metabolismo , Dados de Sequência Molecular , Níquel , Ácido Nitrilotriacético , RNA Ligase (ATP)/genética , RNA Ligase (ATP)/metabolismo , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Sefarose , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...