Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Biochemistry (Mosc) ; 86(9): 1060-1094, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34565312

RESUMO

Viruses exploit the translation machinery of an infected cell to synthesize their proteins. Therefore, viral mRNAs have to compete for ribosomes and translation factors with cellular mRNAs. To succeed, eukaryotic viruses adopt multiple strategies. One is to circumvent the need for m7G-cap through alternative instruments for ribosome recruitment. These include internal ribosome entry sites (IRESs), which make translation independent of the free 5' end, or cap-independent translational enhancers (CITEs), which promote initiation at the uncapped 5' end, even if located in 3' untranslated regions (3' UTRs). Even if a virus uses the canonical cap-dependent ribosome recruitment, it can still perturb conventional ribosomal scanning and start codon selection. The pressure for genome compression often gives rise to internal and overlapping open reading frames. Their translation is initiated through specific mechanisms, such as leaky scanning, 43S sliding, shunting, or coupled termination-reinitiation. Deviations from the canonical initiation reduce the dependence of viral mRNAs on translation initiation factors, thereby providing resistance to antiviral mechanisms and cellular stress responses. Moreover, viruses can gain advantage in a competition for the translational machinery by inactivating individual translational factors and/or replacing them with viral counterparts. Certain viruses even create specialized intracellular "translation factories", which spatially isolate the sites of their protein synthesis from cellular antiviral systems, and increase availability of translational components. However, these virus-specific mechanisms may become the Achilles' heel of a viral life cycle. Thus, better understanding of the unconventional mechanisms of viral mRNA translation initiation provides valuable insight for developing new approaches to antiviral therapy.


Assuntos
Células Eucarióticas/virologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Animais , Células Eucarióticas/fisiologia , Humanos , Sítios Internos de Entrada Ribossomal/fisiologia , RNA Circular/genética , Proteínas Virais/fisiologia
2.
Vet Microbiol ; 255: 109024, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33713975

RESUMO

Senecavirus A (SVA) is classified into the genus Senecavirus in the family Picornaviridae. Its genome is a positive-sense, single-stranded and nonsegmented RNA, approximately 7300 nucleotides in length. A picornaviral genome is essentially an mRNA, which, albeit unmodified with 5' cap structure, can still initiate protein expression by the internal ribosome entry site (IRES). The SVA genome contains a hepatitis C virus-like IRES, in which a pseudoknot structure plays an important role in initiating protein expression. In this study, we constructed a set of SVA (CH-LX-01-2016 strain) minigenomes with all combinations of point mutations in its pseudoknot stem II (PKS-II). The results showed that any combination of point mutations could not significantly interfere with the IRES to initiate protein expression. Further, we constructed a full-length SVA cDNA clone, in which the PKS-II-forming cDNA motif was subjected to site-directed mutagenesis for totally disrupting the PKS-II formation in IRES. Such a modified SVA cDNA clone was transfected into BSR-T7/5 cells, consequently demonstrating that the PKS-II-disrupting IRES interfered neither with protein expression nor with antigenome replication, whereas a competent SVA could not be rescued from the cDNA clone. It was speculated that the mutated motif possibly disrupted a packaging signal for virion assembly, therefore causing the failure of SVA rescue.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , Sítios Internos de Entrada Ribossomal/fisiologia , Picornaviridae , RNA Viral/química , Proteínas Virais/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Cricetinae , DNA Complementar , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Mutação Puntual , RNA Viral/genética , RNA Viral/metabolismo
3.
Cell Rep ; 33(10): 108476, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33296660

RESUMO

Dicistrovirus intergenic region internal ribosomal entry sites (IGR IRESs) do not require initiator tRNA, an AUG codon, or initiation factors and jumpstart translation from the middle of the elongation cycle via formation of IRES/80S complexes resembling the pre-translocation state. eEF2 then translocates the [codon-anticodon]-mimicking pseudoknot I (PKI) from ribosomal A sites to P sites, bringing the first sense codon into the decoding center. Halastavi árva virus (HalV) contains an IGR that is related to previously described IGR IRESs but lacks domain 2, which enables these IRESs to bind to individual 40S ribosomal subunits. By using in vitro reconstitution and cryoelectron microscopy (cryo-EM), we now report that the HalV IGR IRES functions by the simplest initiation mechanism that involves binding to 80S ribosomes such that PKI is placed in the P site, so that the A site contains the first codon that is directly accessible for decoding without prior eEF2-mediated translocation of PKI.


Assuntos
Sítios Internos de Entrada Ribossomal/genética , Iniciação Traducional da Cadeia Peptídica/genética , Vírus de RNA de Cadeia Positiva/genética , Anticódon , Códon/metabolismo , Microscopia Crioeletrônica/métodos , DNA Intergênico/metabolismo , Sítios Internos de Entrada Ribossomal/fisiologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , Fator 2 de Elongação de Peptídeos/metabolismo , Fatores de Iniciação de Peptídeos/genética , Vírus de RNA de Cadeia Positiva/metabolismo , Biossíntese de Proteínas/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , Ribossomos/metabolismo , Replicação Viral/genética , Replicação Viral/fisiologia , Vírus/metabolismo
4.
Nat Commun ; 11(1): 4775, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32963221

RESUMO

Enterovirus 71 (EV71) poses serious threats to human health, particularly in Southeast Asia, and no drugs or vaccines are available. Previous work identified the stem loop II structure of the EV71 internal ribosomal entry site as vital to viral translation and a potential target. After screening an RNA-biased library using a peptide-displacement assay, we identify DMA-135 as a dose-dependent inhibitor of viral translation and replication with no significant toxicity in cell-based studies. Structural, biophysical, and biochemical characterization support an allosteric mechanism in which DMA-135 induces a conformational change in the RNA structure that stabilizes a ternary complex with the AUF1 protein, thus repressing translation. This mechanism is supported by pull-down experiments in cell culture. These detailed studies establish enterovirus RNA structures as promising drug targets while revealing an approach and mechanism of action that should be broadly applicable to functional RNA targeting.


Assuntos
Enterovirus Humano A/genética , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Sítios Internos de Entrada Ribossomal/fisiologia , Replicação Viral/fisiologia , Regiões 5' não Traduzidas , Linhagem Celular , Infecções por Enterovirus/virologia , Regulação Viral da Expressão Gênica , Ribonucleoproteína Nuclear Heterogênea D0/metabolismo , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica , RNA Viral/química , Proteínas Virais/metabolismo
5.
J Virol ; 94(16)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32493820

RESUMO

Foot-and-mouth disease (FMD), which is caused by FMD virus (FMDV), remains a major plague among cloven-hoofed animals worldwide, and its outbreak often has disastrous socioeconomic consequences. A live-attenuated FMDV vaccine will greatly facilitate the global control and eradication of FMD, but a safe and effective attenuated FMDV vaccine has not yet been successfully developed. Here, we found that the internal ribosome entry site (IRES) element in the viral genome is a critical virulence determinant of FMDV, and a nucleotide substitution of cytosine (C) for guanine (G) at position 351 of the IRES endows FMDV with temperature-sensitive and attenuation (ts&att) phenotypes. Furthermore, we demonstrated that the C351G mutation of IRES causes a temperature-dependent translation defect by impairing its binding to cellular pyrimidine tract-binding protein (PTB), resulting in the ts&att phenotypes of FMDV. Natural hosts inoculated with viruses carrying the IRES C351G mutation showed no clinical signs, viremia, virus excretion, or viral transmission but still produced a potent neutralizing antibody response that provided complete protection. Importantly, the IRES C351G mutation is a universal determinant of the ts&att phenotypes of different FMDV strains, and the C351G mutant was incapable of reversion to virulence during in vitro and in vivo passages. Collectively, our findings suggested that manipulation of the IRES, especially its C351G mutation, may serve as a feasible strategy to develop live-attenuated FMDV vaccines.IMPORTANCE The World Organization for Animal Health has called for global control and eradication of foot-and-mouth disease (FMD), the most economically and socially devastating disease affecting animal husbandry worldwide. Live-attenuated vaccines are considered the most effective strategy for prevention, control, and eradication of infectious diseases due to their capacity to induce potent and long-lasting protective immunity. However, efforts to develop FMD virus (FMDV) live-attenuated vaccines have achieved only limited success. Here, by structure-function study of the FMDV internal ribosome entry site (IRES), we find that the C351 mutation of the IRES confers FMDV with an ideal temperature-sensitive attenuation phenotype by decreasing its interaction with cellular pyrimidine tract-binding protein (PTB) to cause IRES-mediated temperature-dependent translation defects. The temperature-sensitive attenuated strains generated by manipulation of the IRES address the challenges of FMDV attenuation differences among various livestock species and immunogenicity maintenance encountered previously, and this strategy can be applied to other viruses with an IRES to rationally design and develop live-attenuated vaccines.


Assuntos
Vírus da Febre Aftosa/genética , Sítios Internos de Entrada Ribossomal/genética , Animais , Anticorpos Neutralizantes/metabolismo , Bovinos , Feminino , Febre Aftosa/virologia , Vírus da Febre Aftosa/metabolismo , Vírus da Febre Aftosa/patogenicidade , Regulação Viral da Expressão Gênica/genética , Sítios Internos de Entrada Ribossomal/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação/genética , Ribossomos/genética , Suínos , Vacinas Atenuadas , Virulência/genética , Replicação Viral/genética
6.
J Virol ; 94(17)2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32581104

RESUMO

Cap-independent translation initiation on picornavirus mRNAs is mediated by an internal ribosomal entry site (IRES) in the 5' untranslated region. The regulation of internal initiation requires the interaction of IRES-transacting factors (ITAFs) with the IRES. In this study, we identified a novel ITAF, heterogeneous nuclear ribonucleoprotein K (hnRNP K), which negatively regulates foot-and-mouth disease virus (FMDV) translation and viral replication. Further investigation revealed that the KH2 and KH3 domains of hnRNP K directly bind to domains II, III, and IV of the FMDV IRES, resulting in the inhibition of IRES-mediated translation by interfering with the recognition of another positive ITAF, polypyrimidine tract-binding protein (PTB). Conversely, hnRNP K-mediated inhibition was antagonized by the viral 3C protease through the cleavage of hnRNP K at the Glu-364 residue during FMDV infection. Interestingly, the N-terminal cleavage product, hnRNP K1-364, retained partial inhibitory effects on IRES activity, whereas the C-terminal cleavage product, hnRNP K364-465, became a positive regulator of FMDV replication. Our findings expand the current understanding of virus-host interactions concerning viral recruitment and the modulation of ITAFs, providing new insights into translational control during viral infection.IMPORTANCE The translation of picornaviral genome RNA mediated by the internal ribosomal entry site (IRES) is a crucial step for virus infections. Virus-host interactions play a critical role in the regulation of IRES-dependent translation, but the regulatory mechanism remains largely unknown. In this study, we identified an ITAF, hnRNP K, that negatively regulates FMDV replication by inhibiting viral IRES-mediated translation. In addition, we describe a novel translational regulation mechanism involving the proteolytic cleavage of hnRNP K by FMDV protease 3C. The cleavage of hnRNP K yields two cleavage products with opposite functions: the cleavage product hnRNP K1-364 retains a partial inhibitory effect on IRES activity, and the cleavage product hnRNP K364-465 becomes a positive regulator of FMDV replication. Our findings shed light on the effect of a novel ITAF on the translational regulation of picornavirus and provide new insights into translational control during viral infection.


Assuntos
Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Sítios Internos de Entrada Ribossomal/fisiologia , Transativadores/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Proteases Virais 3C , Animais , Linhagem Celular , Cricetinae , Vírus da Febre Aftosa/genética , Regulação Viral da Expressão Gênica , Células HEK293 , Humanos , Proteína de Ligação a Regiões Ricas em Polipirimidinas , RNA Mensageiro , Proteínas Virais/genética
7.
Int J Mol Sci ; 21(7)2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32230899

RESUMO

Translation of the hepatitis C virus (HCV) RNA genome is regulated by the internal ribosome entry site (IRES), located in the 5'-untranslated region (5'UTR) and part of the core protein coding sequence, and by the 3'UTR. The 5'UTR has some highly conserved structural regions, while others can assume different conformations. The IRES can bind to the ribosomal 40S subunit with high affinity without any other factors. Nevertheless, IRES activity is modulated by additional cis sequences in the viral genome, including the 3'UTR and the cis-acting replication element (CRE). Canonical translation initiation factors (eIFs) are involved in HCV translation initiation, including eIF3, eIF2, eIF1A, eIF5, and eIF5B. Alternatively, under stress conditions and limited eIF2-Met-tRNAiMet availability, alternative initiation factors such as eIF2D, eIF2A, and eIF5B can substitute for eIF2 to allow HCV translation even when cellular mRNA translation is downregulated. In addition, several IRES trans-acting factors (ITAFs) modulate IRES activity by building large networks of RNA-protein and protein-protein interactions, also connecting 5'- and 3'-ends of the viral RNA. Moreover, some ITAFs can act as RNA chaperones that help to position the viral AUG start codon in the ribosomal 40S subunit entry channel. Finally, the liver-specific microRNA-122 (miR-122) stimulates HCV IRES-dependent translation, most likely by stabilizing a certain structure of the IRES that is required for initiation.


Assuntos
Genoma Viral , Hepacivirus/genética , Hepacivirus/metabolismo , Sítios Internos de Entrada Ribossomal/genética , Biossíntese de Proteínas/fisiologia , RNA Viral/genética , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas , Códon de Iniciação , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 3 em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos , Hepatite C/virologia , Humanos , Sítios Internos de Entrada Ribossomal/fisiologia , MicroRNAs/metabolismo , Modelos Moleculares , Iniciação Traducional da Cadeia Peptídica , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Ribossomos/metabolismo
8.
J Virol ; 94(10)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32161169

RESUMO

Upon infection, the highly structured 5' untranslated region (5' UTR) of picornavirus is involved in viral protein translation and RNA synthesis. As a critical element in the 5' UTR, the internal ribosome entry site (IRES) binds to various cellular proteins to function in the processes of picornavirus replication. Foot-and-mouth disease virus (FMDV) is an important member in the family Picornaviridae, and its 5' UTR contains a functional IRES element. In this study, the cellular heterogeneous nuclear ribonucleoprotein L (hnRNP L) was identified as an IRES-binding protein for FMDV by biotinylated RNA pulldown assays, mass spectrometry (MS) analysis, and determination of hnRNP L-IRES interaction regions. Further, we found that hnRNP L inhibited the growth of FMDV through binding to the viral IRES and that the inhibitory effect of hnRNP L on FMDV growth was not due to FMDV IRES-mediated translation, but to influence on viral RNA synthesis. Finally, hnRNP L was demonstrated to coimmunoprecipitate with RNA-dependent RNA polymerase (3Dpol) in an FMDV RNA-dependent manner in the infected cells. Thus, our results suggest that hnRNP L, as a critical IRES-binding protein, negatively regulates FMDV replication by inhibiting viral RNA synthesis, possibly by remaining in the replication complex.IMPORTANCE Picornaviruses, as a large family of human and animal pathogens, cause a bewildering array of disease syndromes. Many host factors are implicated in the pathogenesis of these viruses, and some proteins interact with the viral IRES elements to affect function. Here, we report for the first time that cellular hnRNP L specifically interacts with the IRES of the picornavirus FMDV and negatively regulates FMDV replication through inhibiting viral RNA synthesis. Further, our results showed that hnRNP L coimmunoprecipitates with FMDV 3Dpol in a viral RNA-dependent manner, suggesting that it may remain in the replication complex to function. The data presented here would facilitate further understanding of virus-host interactions and the pathogenesis of picornavirus infections.


Assuntos
Regiões 5' não Traduzidas , Vírus da Febre Aftosa/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Sítios Internos de Entrada Ribossomal/fisiologia , RNA Viral/biossíntese , Replicação Viral/fisiologia , Animais , Linhagem Celular , Vírus da Febre Aftosa/genética , Regulação Viral da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Imunoprecipitação , Ligação Proteica , RNA Viral/genética , Transcriptoma
9.
Int J Mol Sci ; 21(1)2020 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-31948038

RESUMO

Internal ribosome entry site (IRES)-mediated protein synthesis has been demonstrated to play an important role in resistance to mechanistic target of rapamycin (mTOR) targeted therapies. Previously, we have demonstrated that the IRES trans-acting factor (ITAF), hnRNP A1 is required to promote IRES activity and small molecule inhibitors which bind specifically to this ITAF and curtail IRES activity, leading to mTOR inhibitor sensitivity. Here we report the identification of riluzole (Rilutek®), an FDA-approved drug for amyotrophic lateral sclerosis (ALS), via an in silico docking analysis of FDA-approved compounds, as an inhibitor of hnRNP A1. In a riluzole-bead coupled binding assay and in surface plasmon resonance imaging analyses, riluzole was found to directly bind to hnRNP A1 and inhibited IRES activity via effects on ITAF/RNA-binding. Riluzole also demonstrated synergistic anti-glioblastoma (GBM) affects with mTOR inhibitors in vitro and in GBM xenografts in mice. These data suggest that repurposing riluzole, used in conjunction with mTOR inhibitors, may serve as an effective therapeutic option in glioblastoma.


Assuntos
Antineoplásicos/farmacologia , Glioblastoma/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1/antagonistas & inibidores , Sítios Internos de Entrada Ribossomal/efeitos dos fármacos , Riluzol/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Reposicionamento de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Ribonucleoproteína Nuclear Heterogênea A1/genética , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Humanos , Sítios Internos de Entrada Ribossomal/fisiologia , Camundongos , Camundongos SCID , Simulação de Acoplamento Molecular , Biossíntese de Proteínas/efeitos dos fármacos , Riluzol/química , Riluzol/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores
10.
Elife ; 82019 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-31815666

RESUMO

Hypoxia, a major inducer of angiogenesis, triggers major changes in gene expression at the transcriptional level. Furthermore, under hypoxia, global protein synthesis is blocked while internal ribosome entry sites (IRES) allow specific mRNAs to be translated. Here, we report the transcriptome and translatome signatures of (lymph)angiogenic genes in hypoxic HL-1 mouse cardiomyocytes: most genes are induced at the translatome level, including all IRES-containing mRNAs. Our data reveal activation of (lymph)angiogenic factor mRNA IRESs in early hypoxia. We identify vasohibin1 (VASH1) as an IRES trans-acting factor (ITAF) that is able to bind RNA and to activate the FGF1 IRES in hypoxia, but which tends to inhibit several IRESs in normoxia. VASH1 depletion has a wide impact on the translatome of (lymph)angiogenesis genes, suggesting that this protein can regulate translation positively or negatively in early hypoxia. Translational control thus appears as a pivotal process triggering new vessel formation in ischemic heart.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Hipóxia/metabolismo , Sítios Internos de Entrada Ribossomal/fisiologia , Miócitos Cardíacos/metabolismo , Transativadores/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Humanos , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , RNA Mensageiro/metabolismo , Motivos de Ligação ao RNA , Transcriptoma
11.
J Mol Cell Biol ; 11(10): 911-919, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31504667

RESUMO

Most eukaryotic mRNAs are translated in a cap-dependent fashion; however, under stress conditions, the cap-independent translation driven by internal ribosomal entry sites (IRESs) can serve as an alternative mechanism for protein production. Many IRESs have been discovered from viral or cellular mRNAs to promote ribosome assembly and initiate translation by recruiting different trans-acting factors. Although the mechanisms of translation initiation driven by viral IRESs are relatively well understood, the existence of cellular IRESs is still under debate due to the limitations of translation reporter systems used to assay IRES activities. A recent screen identified > 1000 putative IRESs from viral and human mRNAs, expanding the scope and mechanism for cap-independent translation. Additionally, a large number of circular RNAs lacking free ends were identified in eukaryotic cells, many of which are found to be translated through IRESs. These findings suggest that IRESs may play a previously unappreciated role in driving translation of the new type of mRNA, implying a hidden proteome produced from cap-independent translation.


Assuntos
Sítios Internos de Entrada Ribossomal/fisiologia , Proteoma/metabolismo , Humanos , Sítios Internos de Entrada Ribossomal/genética , Modelos Biológicos , Biossíntese de Proteínas/genética , Biossíntese de Proteínas/fisiologia , Ribossomos/metabolismo
12.
Nat Commun ; 10(1): 3629, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31399592

RESUMO

Picornaviral IRES elements are essential for initiating the cap-independent viral translation. However, three-dimensional structures of these elements remain elusive. Here, we report a 2.84-Å resolution crystal structure of hepatitis A virus IRES domain V (dV) in complex with a synthetic antibody fragment-a crystallization chaperone. The RNA adopts a three-way junction structure, topologically organized by an adenine-rich stem-loop motif. Despite no obvious sequence homology, the dV architecture shows a striking similarity to a circularly permuted form of encephalomyocarditis virus J-K domain, suggesting a conserved strategy for organizing the domain architecture. Recurrence of the motif led us to use homology modeling tools to compute a 3-dimensional structure of the corresponding domain of foot-and-mouth disease virus, revealing an analogous domain organizing motif. The topological conservation observed among these IRESs and other viral domains implicates a structured three-way junction as an architectural scaffold to pre-organize helical domains for recruiting the translation initiation machinery.


Assuntos
Sequência Conservada , Sítios Internos de Entrada Ribossomal/fisiologia , Motivos de Nucleotídeos/fisiologia , Picornaviridae/fisiologia , RNA Viral/química , RNA Viral/fisiologia , Sequência de Bases , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Vírus da Hepatite A , Sítios Internos de Entrada Ribossomal/imunologia , Chaperonas Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Conformação de Ácido Nucleico , Biossíntese de Proteínas , RNA Viral/metabolismo , Ribossomos/química , Ribossomos/metabolismo
13.
Viruses ; 11(7)2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31288421

RESUMO

The 5' untranslated region (UTR) of the RNA genomes of enteroviruses possesses an internal ribosome entry site (IRES) that directs translation of the mRNA by binding to ribosomes. Infection with enterovirus D68 causes respiratory symptoms and is sometimes associated with neurological disorders. The number of reports of the viral infection and neurological disorders has increased in 2010s, although the reason behind this phenomenon remains unelucidated. In this study, we investigated the evolutionary and functional diversity of the 5' UTR of recently circulating strains of the virus. Genomic sequences of 374 viral strains were acquired and subjected to phylogenetic analysis. The IRES activity of the viruses was measured using a luciferase reporter assay. We found a highly conserved sequence in the 5' UTR and also identified the location of variable sites in the predicted RNA secondary structure. IRES activities differed among the strains in some cell lines, including neuronal and respiratory cells, and were especially high in strains of a major lineage from the recent surge. The effect of mutations in the 5' UTR should be studied further in the future for better understanding of viral pathogenesis.


Assuntos
Regiões 5' não Traduzidas/genética , Regiões 5' não Traduzidas/fisiologia , Enterovirus Humano D/genética , Evolução Molecular , Variação Genética , Sítios Internos de Entrada Ribossomal/genética , Linhagem Celular , Enterovirus Humano D/fisiologia , Sítios Internos de Entrada Ribossomal/fisiologia , Conformação de Ácido Nucleico , Filogenia , RNA Mensageiro/metabolismo , RNA Viral/química , RNA Viral/genética , Ribossomos , Proteínas Virais/genética
14.
Open Biol ; 8(11)2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30487301

RESUMO

Beyond the general cap-dependent translation initiation, eukaryotic organisms use alternative mechanisms to initiate protein synthesis. Internal ribosome entry site (IRES) elements are cis-acting RNA regions that promote internal initiation of translation using a cap-independent mechanism. However, their lack of primary sequence and secondary RNA structure conservation, as well as the diversity of host factor requirement to recruit the ribosomal subunits, suggest distinct types of IRES elements. In spite of this heterogeneity, conserved motifs preserve sequences impacting on RNA structure and RNA-protein interactions important for IRES-driven translation. This conservation brings the question of whether IRES elements could consist of basic building blocks, which upon evolutionary selection result in functional elements with different properties. Although RNA-binding proteins (RBPs) perform a crucial role in the assembly of ribonucleoprotein complexes, the versatility and plasticity of RNA molecules, together with their high flexibility and dynamism, determines formation of macromolecular complexes in response to different signals. These properties rely on the presence of short RNA motifs, which operate as modular entities, and suggest that decomposition of IRES elements in short modules could help to understand the different mechanisms driven by these regulatory elements. Here we will review evidence suggesting that model IRES elements consist of the combination of short modules, providing sites of interaction for ribosome subunits, eIFs and RBPs, with implications for definition of criteria to identify novel IRES-like elements genome wide.


Assuntos
Sítios Internos de Entrada Ribossomal/fisiologia , Motivos de Nucleotídeos/fisiologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , Animais , Humanos , Proteínas de Ligação a RNA/metabolismo
15.
Methods Mol Biol ; 1850: 289-308, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30242694

RESUMO

We present an alternative production platform for the synthesis of complex proteins. Apart from conventionally applied protein production using engineered mammalian cell lines, this protocol describes the preparation and principle of cell-free protein synthesis systems based on CHO cell lysates. The CHO cell-free system contains endogenous microsomes derived from the endoplasmic reticulum, which enables a direct integration of membrane proteins into a nature like milieu and the introduction of posttranslational modifications. Different steps of system development are described including the cultivation of CHO cells, cell harvesting and cell disruption to prepare translationally active CHO cell lysates. The requirements for DNA templates and the generation of linear DNA templates suitable for the CHO cell-free reaction is further depicted to underline the opportunity to produce different protein variants in a short period. This experimental setup provides a basis for high-throughput applications. The productivity of the CHO cell-free systems is further increased by using a non-canonical translation initiation due to the attachment of an internal ribosomal entry site of the Cricket paralysis virus (CRPV IRES) to the 5´ UTR of the desired gene. In this way, a direct interaction of the IRES structure with the ribosome facilitates a translation factor independent initiation of translation. Cell-free reactions were performed in fast and efficient batch reactions leading to protein yields up to 40 µg/mL. The reaction format was further adjusted to a continuous exchange CHO cell-free reaction (CHO CECF) to prolong reaction time and thereby increase the productivity of the cell-free systems. Finally, protein yields up to 1 g/L were obtained. The CHO CECF system represents a sophisticated resource to address structural and functional aspects of difficult-to-express proteins in fundamental and applied research.


Assuntos
Sistema Livre de Células/metabolismo , Biossíntese de Proteínas/fisiologia , Animais , Biotecnologia , Células CHO , Cricetinae , Cricetulus , Sítios Internos de Entrada Ribossomal/genética , Sítios Internos de Entrada Ribossomal/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Biossíntese de Proteínas/genética , Processamento de Proteína Pós-Traducional/genética , Processamento de Proteína Pós-Traducional/fisiologia
16.
Mol Cell ; 71(3): 364-374, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-30075139

RESUMO

The ribosome has recently transitioned from being viewed as a passive, indiscriminate machine to a more dynamic macromolecular complex with specialized roles in the cell. Here, we discuss the historical milestones from the discovery of the ribosome itself to how this ancient machinery has gained newfound appreciation as a more regulatory participant in the central dogma of gene expression. The first emerging examples of direct changes in ribosome composition at the RNA and protein level, coupled with an increased awareness of the role individual ribosomal components play in the translation of specific mRNAs, is opening a new field of study centered on ribosome-mediated control of gene regulation. In this Perspective, we discuss our current understanding of the known functions for ribosome heterogeneity, including specialized translation of individual transcripts, and its implications for the regulation and expression of key gene regulatory networks. In addition, we suggest what the crucial next steps are to ascertain the extent of ribosome heterogeneity and specialization and its importance for regulation of the proteome within subcellular space, across different cell types, and during multi-cellular organismal development.


Assuntos
Ribossomos/metabolismo , Ribossomos/fisiologia , Animais , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Sítios Internos de Entrada Ribossomal/fisiologia , Biossíntese de Proteínas , RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas Ribossômicas/metabolismo
17.
Vaccine ; 36(32 Pt B): 4846-4850, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-30037477

RESUMO

Newcastle disease virus (NDV) has been used as a vector in the development of vaccines and gene therapy. A majority of these NDV vectors express only a single foreign gene through either an independent transcription unit (ITU) or an internal ribosomal entry site (IRES). In the present study, we combined the ITU and IRES methods to generate a novel NDV LaSota strain-based recombinant virus vectoring the red fluorescence protein (RFP) and the green fluorescence protein (GFP) genes. Biological assessments of the recombinant virus, rLS/IRES-RFP/GFP, showed that it was slightly attenuated in vivo, yet maintained similar growth dynamics and viral yields in vitro when compared to the parental LaSota virus. Expression of both the RFP and GFP was detected from the rLS/IRES-RFP/GFP virus-infected DF-1 cells by fluorescence microscopy. These data suggest that the rLS/IRES-RFP/GFP virus may be used as a multivalent vector for the development of vaccines and gene therapy agents.


Assuntos
Doença de Newcastle/genética , Vírus da Doença de Newcastle/genética , Animais , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Sítios Internos de Entrada Ribossomal/genética , Sítios Internos de Entrada Ribossomal/fisiologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Doença de Newcastle/imunologia , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/imunologia , Vacinas Virais/uso terapêutico , Proteína Vermelha Fluorescente
18.
Cerebellum ; 17(1): 72-77, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29374372

RESUMO

We have discovered that the P/Q-type voltage-gated Ca2+ channel (VGCC) gene, CACNA1A, encodes both the α1A (Cav2.1) subunit and a newly recognized transcription factor, α1ACT, by means of a novel internal ribosomal entry site (IRES) within the α1A C-terminal coding region. α1ACT, when mutated with an expansion of the polyglutamine tract in the C-terminus, gives rise to spinocerebellar ataxia type 6 (SCA6). Because silencing of the entire CACNA1A gene would result in the loss of the essential Cav2.1 channel, the IRES controlling α1ACT expression is an excellent target for selective silencing of α1ACT as a therapeutic intervention for SCA6. We performed a high-throughput screen of FDA-approved small molecules using a dual luciferase reporter system and identified ten hits able to selectively inhibit the IRES. We identified four main candidates that showed selective suppression of α1ACT relative to α1A in HEK cells expressing a native CACNA1A vector. We previously pursued another avenue of molecular intervention through miRNA silencing. We studied three human miRNAs (miRNA-711, -3191-5p, -4786) that would potentially bind to sequences within the CACNA1A IRES region, based on an miRNA prediction program. Only miRNA-3191-5p was found to selectively inhibit the translation of α1ACT in cells. We developed a hyperacute model of SCA6 in mice by injecting a pathogenic form of the IRES-mediated α1ACT (AAV9-α1ACTQ33). Finally, we tested the effectiveness of the miRNA therapy by co-expressing either control miRNA or miRNA-3191-5p and found that miRNA-3191-5p decreased the levels of α1ACTQ33 and prevented the hyperacute disease in mice. These studies provide the proof of principle that a therapy directed at selectively preventing α1ACT expression could be used to treat SCA6.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio/metabolismo , Regulação da Expressão Gênica/fisiologia , Sítios Internos de Entrada Ribossomal/fisiologia , Ataxias Espinocerebelares/tratamento farmacológico , Ajmalina/farmacologia , Animais , Canais de Cálcio/genética , Canais de Cálcio Tipo L/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Concentração Inibidora 50 , Sítios Internos de Entrada Ribossomal/efeitos dos fármacos , MicroRNAs/genética , MicroRNAs/metabolismo , Mutação/genética , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo , Ataxias Espinocerebelares/genética , Transfecção , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia
19.
J Virol ; 92(2)2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29093099

RESUMO

All viruses must successfully harness the host translational apparatus and divert it towards viral protein synthesis. Dicistroviruses use an unusual internal ribosome entry site (IRES) mechanism whereby the IRES adopts a three-pseudoknot structure that accesses the ribosome tRNA binding sites to directly recruit the ribosome and initiate translation from a non-AUG start site. A subset of dicistroviruses, including the honey bee Israeli acute paralysis virus (IAPV), encode an extra stem-loop (SLVI) 5' -adjacent to the IGR IRES. Previously, the function of this additional stem-loop is unknown. Here, we provide mechanistic and functional insights into the role of SLVI in IGR IRES translation and in virus infection. Biochemical analyses of a series of mutant IRESs demonstrated that SLVI does not function in ribosome recruitment but is required for proper ribosome positioning on the IRES to direct translation. Using a chimeric infectious clone derived from the related Cricket paralysis virus, we showed that the integrity of SLVI is important for optimal viral translation and viral yield. Based on structural models of ribosome-IGR IRES complexes, the SLVI is predicted to be in the vicinity of the ribosome E site. We propose that SLVI of IAPV IGR IRES functionally mimics interactions of an E-site tRNA with the ribosome to direct positioning of the tRNA-like domain of the IRES in the A site.IMPORTANCEViral internal ribosome entry sites are RNA elements and structures that allow some positive-sense monopartite RNA viruses to hijack the host ribosome to start viral protein synthesis. We demonstrate that a unique stem-loop structure is essential for optimal viral protein synthesis and for virus infection. Biochemical evidence shows that this viral stem-loop RNA structure impacts a fundamental property of the ribosome to start protein synthesis.


Assuntos
Abelhas/virologia , Dicistroviridae/genética , Sítios Internos de Entrada Ribossomal/fisiologia , Biossíntese de Proteínas , Ribossomos/química , Animais , DNA Intergênico/genética , Dicistroviridae/química , Dicistroviridae/metabolismo , Drosophila/genética , Mutação , RNA de Transferência/fisiologia , Ribossomos/genética , Ribossomos/metabolismo
20.
Int J Biochem Cell Biol ; 93: 46-51, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29107182

RESUMO

Many studies demonstrated that there are several type bands of prion protein in cells. However, the formation of different prion protein bands is elusive. After several low molecular weight bands of prion protein appeared in SMB-S15 cells infected with scrapie agent Chandler, we think that IRES-dependent translation mechanism induced by prion is involved in the formation of prion protein bands. Then we designed a series of pPrP-GFP fusing plasmids and bicistronic plasmids to identify the IRES sites of prion protein gene and found 3 IRES sites inside of PrP mRNA. We also demonstrated that cap-independent translation of PrP was associated with the ER stress through Tunicamycin treatment. We still found that only IRE1 and PERK pathway regulated the IRES-dependent translation of PrP in this study. Our results indicated, we found that PrP gene had an IRES-dependent translation initiation mechanism and we successfully identified the IRESs inside of the prion protein gene.


Assuntos
Sítios Internos de Entrada Ribossomal/fisiologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , Proteínas Priônicas/biossíntese , RNA Mensageiro/metabolismo , Animais , Cricetinae , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Células HEK293 , Humanos , Proteínas Priônicas/genética , RNA Mensageiro/genética , Tunicamicina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA