Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 195: 106488, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38565397

RESUMO

Given their highly polarized morphology and functional singularity, neurons require precise spatial and temporal control of protein synthesis. Alterations in protein translation have been implicated in the development and progression of a wide range of neurological and neurodegenerative disorders, including Huntington's disease (HD). In this study we examined the architecture of polysomes in their native brain context in striatal tissue from the zQ175 knock-in mouse model of HD. We performed 3D electron tomography of high-pressure frozen and freeze-substituted striatal tissue from HD models and corresponding controls at different ages. Electron tomography results revealed progressive remodelling towards a more compacted polysomal architecture in the mouse model, an effect that coincided with the emergence and progression of HD related symptoms. The aberrant polysomal architecture is compatible with ribosome stalling phenomena. In fact, we also detected in the zQ175 model an increase in the striatal expression of the stalling relief factor EIF5A2 and an increase in the accumulation of eIF5A1, eIF5A2 and hypusinated eIF5A1, the active form of eIF5A1. Polysomal sedimentation gradients showed differences in the relative accumulation of 40S ribosomal subunits and in polysomal distribution in striatal samples of the zQ175 model. These findings indicate that changes in the architecture of the protein synthesis machinery may underlie translational alterations associated with HD, opening new avenues for understanding the progression of the disease.

2.
Mol Cell ; 84(8): 1527-1540.e7, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38521064

RESUMO

Nucleolar stress (NS) has been associated with age-related diseases such as cancer or neurodegeneration. To investigate how NS triggers toxicity, we used (PR)n arginine-rich peptides present in some neurodegenerative diseases as inducers of this perturbation. We here reveal that whereas (PR)n expression leads to a decrease in translation, this occurs concomitant with an accumulation of free ribosomal (r) proteins. Conversely, (PR)n-resistant cells have lower rates of r-protein synthesis, and targeting ribosome biogenesis by mTOR inhibition or MYC depletion alleviates (PR)n toxicity in vitro. In mice, systemic expression of (PR)97 drives widespread NS and accelerated aging, which is alleviated by rapamycin. Notably, the generalized accumulation of orphan r-proteins is a common outcome of chemical or genetic perturbations that induce NS. Together, our study presents a general model to explain how NS induces cellular toxicity and provides in vivo evidence supporting a role for NS as a driver of aging in mammals.


Assuntos
Neoplasias , Ribossomos , Camundongos , Animais , Ribossomos/metabolismo , Envelhecimento/genética , Peptídeos/metabolismo , Sirolimo/farmacologia , Neoplasias/metabolismo , Nucléolo Celular/genética , Mamíferos
3.
Viruses ; 16(2)2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38399981

RESUMO

Alphaviruses can replicate in arthropods and in many vertebrate species including humankind, but only in vertebrate cells do infections with these viruses result in a strong inhibition of host translation and transcription. Translation shutoff by alphaviruses is a multifactorial process that involves both host- and virus-induced mechanisms, and some of them are not completely understood. Alphavirus genomes contain cis-acting elements (RNA structures and dinucleotide composition) and encode protein activities that promote the translational and transcriptional resistance to type I IFN-induced antiviral effectors. Among them, IFIT1, ZAP and PKR have played a relevant role in alphavirus evolution, since they have promoted the emergence of multiple viral evasion mechanisms at the translational level. In this review, we will discuss how the adaptations of alphaviruses to vertebrate hosts likely involved the acquisition of new features in viral mRNAs and proteins to overcome the effect of type I IFN.


Assuntos
Alphavirus , Interferon Tipo I , Animais , Alphavirus/fisiologia , Linhagem Celular , Interferon Tipo I/genética , Vertebrados , Tropismo , Antivirais/farmacologia , Replicação Viral
4.
EMBO J ; 40(13): e103311, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33978236

RESUMO

Due to their capability to transport chemicals or proteins into target cells, cell-penetrating peptides (CPPs) are being developed as therapy delivery tools. However, and despite their interesting properties, arginine-rich CPPs often show toxicity for reasons that remain poorly understood. Using a (PR)n dipeptide repeat that has been linked to amyotrophic lateral sclerosis (ALS) as a model of an arginine-rich CPP, we here show that the presence of (PR)n leads to a generalized displacement of RNA- and DNA-binding proteins from chromatin and mRNA. Accordingly, any reaction involving nucleic acids, such as RNA transcription, translation, splicing and degradation, or DNA replication and repair, is impaired by the presence of the CPPs. Interestingly, the effects of (PR)n are fully mimicked by protamine, a small arginine-rich protein that displaces histones from chromatin during spermatogenesis. We propose that widespread coating of nucleic acids and consequent displacement of RNA- and DNA-binding factors from chromatin and mRNA accounts for the toxicity of arginine-rich CPPs, including those that have been recently associated with the onset of ALS.


Assuntos
Arginina/genética , Peptídeos Penetradores de Células/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a RNA/genética , Esclerose Amiotrófica Lateral/genética , Linhagem Celular Tumoral , Cromatina/genética , DNA/genética , Células HeLa , Histonas/genética , Humanos , Ácidos Nucleicos/genética , RNA/genética , Splicing de RNA/genética , RNA Mensageiro/genética , Espermatogênese/genética
5.
J Virol ; 94(3)2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31723025

RESUMO

Alphaviruses are insect-borne viruses that alternate between replication in mosquitoes and vertebrate species. Adaptation of some alphaviruses to vertebrate hosts has involved the acquisition of an RNA structure (downstream loop [DLP]) in viral subgenomic mRNAs that confers translational resistance to protein kinase (PKR)-mediated eIF2α phosphorylation. Here, we found that, in addition to promoting eIF2-independent translation of viral subgenomic mRNAs, presence of the DLP structure also increased the resistance of alphavirus to type I interferon (IFN). Aura virus (AURAV), an ecologically isolated relative of Sindbis virus (SV) that is poorly adapted to replication in vertebrate cells, displayed a nonfunctional DLP structure and dramatic sensitivity to type I IFN. Our data suggest that an increased resistance to IFN emerged during translational adaptation of alphavirus mRNA to vertebrate hosts, reinforcing the role that double-stranded RNA (dsRNA)-activated protein kinase (PKR) plays as both a constitutive and IFN-induced antiviral effector. Interestingly, a mutant SV lacking the DLP structure (SV-ΔDLP) and AURAV both showed a marked oncotropism for certain tumor cell lines that have defects in PKR expression and/or activation. AURAV selectively replicated in and killed some cell lines derived from human hepatocarcinoma (HCC) that lacked PKR response to infection or poly(I·C) transfection. The oncolytic activities of SV-ΔDLP and AURAV were also confirmed using tumor xenografts in mice, showing tumor regression activities comparable to wild-type SV. Our data show that translation of alphavirus subgenomic mRNAs plays a central role in IFN susceptibility and cell tropism, suggesting an unanticipated oncolytic potential that some naive arboviruses may have in virotherapy.IMPORTANCE Interferons (IFNs) induce the expression of a number of antiviral genes that protect the cells of vertebrates against viruses and other microbes. The susceptibility of cells to viruses greatly depends on the level and activity of these antiviral effectors but also on the ability of viruses to counteract this antiviral response. Here, we found that the level of one of the main IFN effectors in the cell, the dsRNA-activated protein kinase (PKR), greatly determines the permissiveness of cells to alphaviruses that lack mechanisms to counteract its activation. These naive viruses also showed a hypersensitivity to IFN, suggesting that acquisition of IFN resistance (even partial) has probably been involved in expanding the host range of alphaviruses in the past. Interestingly, some of these naive viruses showed a marked oncotropism for some tumor cell lines derived from human hepatocarcinoma (HCC), opening the possibility of their use in oncolytic therapy to treat human tumors.


Assuntos
Alphavirus/genética , Alphavirus/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Interferons/metabolismo , Proteínas Quinases/metabolismo , RNA de Cadeia Dupla/metabolismo , Animais , Antivirais/farmacologia , Apoptose , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Hepáticas , Camundongos SCID , Fosforilação , RNA Mensageiro/metabolismo , Vírus Sindbis/genética , Vertebrados/genética , Replicação Viral/efeitos dos fármacos
6.
Bio Protoc ; 10(16): e3713, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-33659377

RESUMO

The nucleotides involved in RNA-RNA interaction can be tagged by chemical- or UV-induced crosslinking, and further identified by classical or modern high throughput techniques. The contacts of mRNA with 18S rRNA that occur along the mRNA channel of 40S subunit have been mapped by site-specific UV crosslinking followed by reverse transcriptase termination sites (RTTS) using radioactive or fluorescent oligonucleotides. However, the sensitivity of this technique is restricted to the detection of those fragments that resulted from the most frequent crosslinkings. Here, we combined RTTS with RNAseq to map the mRNA-18S rRNA contacts with a much deeper resolution. Although aimed to detect the interaction of mRNA with the ES6S region of 18S rRNA, this technique can also be applied to map the interaction of mRNA with other non-coding RNA molecules (e.g., snRNAs, microRNAs and lncRNAs) during transcription, splicing or RNA-mediated postranscriptional regulation.

7.
Elife ; 82019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31789591

RESUMO

Loading of mRNA onto the ribosomal 43S pre-initiation complex (PIC) and its subsequent scanning require the removal of the secondary structure of the by RNA helicases such as eIF4A. However, the topology and mechanics of the scanning complex bound to mRNA (48S-PIC) and the influence of its solvent-side composition on the scanning process are poorly known. Here, we found that the ES6S region of the 48S-PIC constitutes an extended binding channel for eIF4A-mediated unwinding of mRNA and scanning. Blocking ES6S inhibited the cap-dependent translation of mRNAs that have structured 5' UTRs (including G-quadruplexes), many of which are involved in signal transduction and growth, but it did not affect IRES-driven translation. Genome-wide analysis of mRNA translation revealed a great diversity in ES6S-mediated scanning dependency. Our data suggest that mRNA threading into the ES6S region makes scanning by 48S PIC slower but more processive. Hence, we propose a topological and functional model of the scanning 48S-PIC.


Assuntos
Fator de Iniciação 4F em Eucariotos/genética , RNA Helicases/química , RNA Mensageiro/genética , Proteínas de Saccharomyces cerevisiae/genética , Transdução Genética , Regiões 5' não Traduzidas/genética , Quadruplex G , Iniciação Traducional da Cadeia Peptídica/genética , Biossíntese de Proteínas/genética , RNA Helicases/genética , RNA Mensageiro/química , Ribossomos/genética , Saccharomyces cerevisiae/genética
8.
Nucleic Acids Res ; 46(8): 4176-4187, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29415133

RESUMO

The topology and dynamics of the scanning ribosomal 43S pre-initiation complex (PIC) bound to mRNA and initiation factors (eIFs) are probably the least understood aspects of translation initiation in eukaryotes. Recently, we described a trapping mechanism in alphavirus that stalls the PIC during scanning of viral mRNA. Using this model, we were able to snapshot for the first time the eIF4A helicase bound to mRNA in a 48S initiation complex assembled in vitro. This interaction was only detected in the presence of the natural stem loop structure (DLP) located downstream from the AUG in viral mRNA that promoted stalling of the PIC, suggesting that DLP stability was enough to jam the helicase activity of eIF4A in a fraction of assembled 48S complexes. However, a substantial proportion of DLP mRNA molecules were effectively unwound by eIF4A in vitro, an activity that alphaviruses counteract in infected cells by excluding eIF4A from viral factories. Our data indicated that eIF4A-mRNA contact occurred in (or near) the ES6S region of the 40S subunit, suggesting that incoming mRNA sequences penetrate through the ES6S region during the scanning process. We propose a topological model of the scanning PIC and how some viruses have exploited this topology to translate their mRNAs with fewer eIF requirements.


Assuntos
Alphavirus/genética , Fator de Iniciação 4A em Eucariotos/química , Iniciação Traducional da Cadeia Peptídica , RNA Mensageiro/química , RNA Viral/química , Subunidades Ribossômicas Menores de Eucariotos/química , Animais , Linhagem Celular , Fator de Iniciação 4A em Eucariotos/metabolismo , Modelos Moleculares , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo
9.
RNA Biol ; 13(12): 1223-1227, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27824302

RESUMO

Location of the translation initiation codon generally requires scanning of the 43S ribosomal preinitiation complex (43S PIC) from the 5' of the mRNA. Associated RNA helicases can facilitate movement of the 43S PIC by removing secondary structure present in the 5' UTR of mRNA, which is required for codon inspection. The canonical RNA-dependent helicase eIF4A is directly involved in this process, as part of the eIF4F complex (eIF4G + eIF4A + eIF4E) that associates first with mRNA and then recruits the 43S PIC to initiate scanning. The topology and operational mechanism of the scanning PIC are probably the least understood aspects of the initiation step. Recent findings from translation of alphavirus mRNA, together with new biochemical and structural data of the 43S PIC, suggest a role for the ES6S region of 40S as the gateway for mRNA entry during scanning. The presence of eIF4G-eIF4A complex in this region, interacting with the incoming mRNA, supports a model where eIF4A could work ahead of the scanning complex during translation initiation. Here we present additional data supporting this model.


Assuntos
RNA Mensageiro/química , Ribossomos/metabolismo , Proteínas Virais/metabolismo , Vírus/metabolismo , Fator de Iniciação 4A em Eucariotos/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Fator de Iniciação 4G em Eucariotos/metabolismo , Modelos Moleculares , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Viral/química , RNA Viral/genética , Vírus/genética
10.
Nucleic Acids Res ; 44(9): 4368-80, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-26984530

RESUMO

During translation initiation, eukaryotic initiation factor 2 (eIF2) delivers the Met-tRNA to the 40S ribosomal subunit to locate the initiation codon (AUGi) of mRNA during the scanning process. Stress-induced eIF2 phosphorylation leads to a general blockade of translation initiation and represents a key antiviral pathway in mammals. However, some viral mRNAs can initiate translation in the presence of phosphorylated eIF2 via stable RNA stem-loop structures (DLP; Downstream LooP) located in their coding sequence (CDS), which promote 43S preinitiation complex stalling on the initiation codon. We show here that during the scanning process, DLPs of Alphavirus mRNA become trapped in ES6S region (680-914 nt) of 18S rRNA that are projected from the solvent side of 40S subunit. This trapping can lock the progress of the 40S subunit on the mRNA in a way that places the upstream initiator AUGi on the P site of 40S subunit, obviating the participation of eIF2. Notably, the DLP structure is released from 18S rRNA upon 60S ribosomal subunit joining, suggesting conformational changes in ES6Ss during the initiation process. These novel findings illustrate how viral mRNA is threaded into the 40S subunit during the scanning process, exploiting the topology of the 40S subunit solvent side to enhance its translation in vertebrate hosts.


Assuntos
Alphavirus/genética , Iniciação Traducional da Cadeia Peptídica , RNA Mensageiro/genética , RNA Viral/genética , Aedes , Alphavirus/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Códon de Iniciação , Cricetinae , Regulação Viral da Expressão Gênica , Sequências Repetidas Invertidas , Modelos Moleculares , Estabilidade de RNA , RNA Mensageiro/química , RNA Mensageiro/metabolismo , RNA Ribossômico 18S/química , RNA Ribossômico 18S/fisiologia , RNA Viral/química , RNA Viral/metabolismo , Ribossomos/fisiologia
11.
PLoS One ; 7(10): e47272, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23110064

RESUMO

The reversible phosphorylation of the alpha-subunit of eukaryotic translation initiation factor 2 (eIF2alpha) is a well-characterized mechanism of translational control in response to a wide variety of cellular stresses, including viral infection. Beside PKR, the eIF2alpha kinase GCN2 participates in the cellular response against viral infection by RNA viruses with central nervous system tropism. PKR has also been involved in the antiviral response against HIV-1, although this antiviral effect is very limited due to the distinct mechanisms evolved by the virus to counteract PKR action. Here we report that infection of human cells with HIV-1 conveys the proteolytic cleavage of GCN2 and that purified HIV-1 and HIV-2 proteases produce direct proteolysis of GCN2 in vitro, abrogating the activation of GCN2 by HIV-1 RNA. Transfection of distinct cell lines with a plasmid encoding an HIV-1 cDNA clone competent for a single round of replication resulted in the activation of GCN2 and the subsequent eIF2alpha phosphorylation. Moreover, transfection of GCN2 knockout cells or cells with low levels of phosphorylated eIF2alpha with the same HIV-1 cDNA clone resulted in a marked increase of HIV-1 protein synthesis. Also, the over-expression of GCN2 in cells led to a diminished viral protein synthesis. These findings suggest that viral RNA produced during HIV-1 infection activates GCN2 leading to inhibition of viral RNA translation, and that HIV-1 protease cleaves GCN2 to overcome its antiviral effect.


Assuntos
HIV-1/metabolismo , HIV-1/patogenicidade , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/biossíntese , Animais , Western Blotting , Células COS , Linhagem Celular , Chlorocebus aethiops , Fator de Iniciação 2 em Eucariotos/metabolismo , HIV-1/genética , Células HeLa , Humanos , Proteínas Serina-Treonina Quinases/genética , RNA Viral/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Virais/genética
12.
J Virol ; 86(17): 9484-94, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761388

RESUMO

Members of the Alphavirus genus are arboviruses that alternate replication in mosquitoes and vertebrate hosts. In vertebrate cells, the alphavirus resists the activation of antiviral RNA-activated protein kinase (PKR) by the presence of a prominent RNA structure (downstream loop [DLP]) located in viral 26S transcripts, which allows an eIF2-independent translation initiation of these mRNAs. This article shows that DLP structure is essential for replication of Sindbis virus (SINV) in vertebrate cell lines and animals but is dispensable for replication in insect cells, where no ortholog of the vertebrate PKR gene has been found. Sequence comparisons and structural RNA analysis revealed the evolutionary conservation of DLP in SINV and predicted the existence of equivalent DLP structures in many members of the Alphavirus genus. A mutant SINV lacking the DLP structure evolved in murine cells to recover a wild-type phenotype by creating an alternative structure in the RNA that restored the translational independence for eIF2. Genetic, phylogenetic, and biochemical data presented here support an evolutionary scenario for the natural history of alphaviruses, in which the acquisition of DLP structure in their mRNAs probably allowed the colonization of vertebrate host and the consequent geographic expansion of some of these viruses worldwide.


Assuntos
Alphavirus/crescimento & desenvolvimento , Alphavirus/genética , Biossíntese de Proteínas , RNA Viral/genética , Vertebrados/virologia , Adaptação Fisiológica , Alphavirus/química , Alphavirus/classificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Evolução Molecular , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Sequências Repetidas Invertidas , Camundongos , Dados de Sequência Molecular , Mariposas , Conformação de Ácido Nucleico , Filogenia , RNA Viral/química , RNA Viral/metabolismo , Replicação Viral
13.
PLoS One ; 7(5): e35915, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22574127

RESUMO

In this work we have described the translatome of two mammalian cell lines, NIH3T3 and Jurkat, by scoring the relative polysome association of ∼10,000 mRNA under normal and ER stress conditions. We have found that translation efficiencies of mRNA correlated poorly with transcript abundance, although a general tendency was observed so that the highest translation efficiencies were found in abundant mRNA. Despite the differences found between mouse (NIH3T3) and human (Jurkat) cells, both cell types share a common translatome composed by ∼800-900 mRNA that encode proteins involved in basic cellular functions. Upon stress, an extensive remodeling in translatomes was observed so that translation of ∼50% of mRNA was inhibited in both cell types, this effect being more dramatic for those mRNA that accounted for most of the cell translation. Interestingly, we found two subsets comprising 1000-1500 mRNA whose translation resisted or was induced by stress. Translation arrest resistant class includes many mRNA encoding aminoacyl tRNA synthetases, ATPases and enzymes involved in DNA replication and stress response such as BiP. This class of mRNA is characterized by high translation rates in both control and stress conditions. Translation inducible class includes mRNA whose translation was relieved after stress, showing a high enrichment in early response transcription factors of bZIP and zinc finger C2H2 classes. Unlike yeast, a general coordination between changes in translation and transcription upon stress (potentiation) was not observed in mammalian cells. Among the different features of mRNA analyzed, we found a relevant association of translation efficiency with the presence of upstream ATG in the 5'UTR and with the length of coding sequence of mRNA, and a looser association with other parameters such as the length and the G+C content of 5'UTR. A model for translatome remodeling during the acute phase of stress response in mammalian cells is proposed.


Assuntos
Estresse do Retículo Endoplasmático/genética , Biossíntese de Proteínas , Animais , Humanos , Células Jurkat , Camundongos , Células NIH 3T3 , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Transcrição Gênica
14.
PLoS One ; 6(2): e16711, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21311764

RESUMO

Most viruses express during infection products that prevent or neutralize the effect of the host dsRNA activated protein kinase (PKR). Translation of Sindbis virus (SINV) mRNA escapes to PKR activation and eIF2 phosphorylation in infected cells by a mechanism that requires a stem loop structure in viral 26S mRNA termed DLP to initiate translation in the absence of functional eIF2. Unlike the rest of viruses tested, we found that Alphavirus infection allowed a strong PKR activation and eIF2α phosphorylation in vitro and in infected animals so that the presence of DLP structure in mRNA was critical for translation and replication of SINV. Interestingly, infection of MEFs with some viruses that express PKR inhibitors prevented eIF2α phosphorylation after superinfection with SINV, suggesting that viral anti-PKR mechanisms could be exchangeable. Thus, translation of SINV mutant lacking the DLP structure (ΔDLP) in 26S mRNA was partially rescued in cells expressing vaccinia virus (VV) E3 protein, a known inhibitor of PKR. This case of heterotypic complementation among evolutionary distant viruses confirmed experimentally a remarkable case of convergent evolution in viral anti-PKR mechanisms. Our data reinforce the critical role of PKR in regulating virus-host interaction and reveal the versatility of viruses to find different solutions to solve the same conflict.


Assuntos
Evolução Molecular , Variação Genética , Interações Hospedeiro-Patógeno/genética , Vírus/genética , eIF-2 Quinase/antagonistas & inibidores , Animais , Sequência de Bases , Células Cultivadas , Variação Genética/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Transdução de Sinais/genética , Vírus Sindbis/genética , Vírus Sindbis/metabolismo , Replicação Viral/genética , Vírus/metabolismo , Vírus/patogenicidade , eIF-2 Quinase/genética
15.
Proc Natl Acad Sci U S A ; 107(21): 9837-42, 2010 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-20457920

RESUMO

Infection of cultured cells with lytic animal viruses often results in the selective inhibition of host protein synthesis, whereas viral mRNA is efficiently translated under these circumstances. This phenomenon, known as "shut off," has been well described at the molecular level for some viruses, but there is not yet any direct or indirect evidence supporting the idea that it also should operate in animals infected with viruses. To address this issue, we constructed recombinant Sindbis virus (SV)-expressing reporter mRNA, the translation of which is sensitive or resistant to virus-induced shut off. As found in cultured cells, replication of SV in mouse brain was associated with a strong phosphorylation of eukaryotic initiation factor (eIF2) that prevented translation of reporter mRNA (luciferase and EGFP). Translation of these reporters was restored in vitro, in vivo, and ex vivo when a viral RNA structure, termed downstream hairpin loop, present in viral 26S mRNA, was placed at the 5' end of reporter mRNAs. By comparing the expression of shut off-sensitive and -resistant reporters, we unequivocally concluded that replication of SV in animal tissues is associated with a profound inhibition of nonviral mRNA translation. A strategy as simple as that followed here might be applicable to other viruses to evaluate their interference on host translation in infected animals.


Assuntos
Infecções por Alphavirus/virologia , Biossíntese de Proteínas , Vírus Sindbis/fisiologia , Proteínas Virais/genética , Animais , Encéfalo/virologia , Linhagem Celular , Cricetinae , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Genes Reporter , Camundongos , RNA Mensageiro/genética , Ratos , Vírus Sindbis/genética , Replicação Viral
16.
J Virol ; 84(10): 5043-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20219905

RESUMO

The relevance of translational control in the gene expression and oncotropism of the autonomous parvoviruses was investigated with MVMp, the prototype strain of minute virus of mice (MVM), infecting normal and transformed rodent and human cells of different tissue origins. Mouse embryo fibroblasts (MEFs) and NIH 3T3 fibroblasts were resistant to MVMp infection, but 3T3 fibroblasts derived from double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) knockout mice (PKR(o/o)) behaved in a manner that was highly permissive to productive MVMp replication. NIH 3T3 resistance correlated with significant phosphorylation of eukaryotic translation initiation factor 2 (eIF2) occurring at early time points after infection. Permissive PKR(o/o) cells were converted to MVMp-restrictive cells after reintroduction of the PKR gene by transfection. Conversely, regulated expression of the vaccinia virus E3 protein, a PKR inhibitor, in MEFs prevented eIF2alpha phosphorylation and increased MVMp protein synthesis. In vitro-synthesized genome-length R1 mRNA of MVMp was a potent activator of PKR. Virus-resistant primary MEFs and NIH 3T3 cells responded to MVMp infection with significant increases in eIF2alpha phosphorylation. In contrast, virus-permissive mouse (PKR(o/o), BHK21, and A9) and human transformed (NB324K fibroblast, U373 glioma, and HepG2 hepatoma) cells consistently showed no significant increase in the level of eIF2alpha phosphorylation following MVMp infection. The synthesis of the viral NS1 protein was inversely correlated with the steady-state PKR levels. Our results show that the PKR-mediated antiviral response is an important mechanism for control of productive MVMp infection, and its impairment in human transformed cells allowed efficient MVMp gene expression. PKR translational control may therefore contribute to the oncolysis of MVMp and other autonomous parvoviruses.


Assuntos
Vírus Miúdo do Camundongo/imunologia , Vírus Miúdo do Camundongo/patogenicidade , Biossíntese de Proteínas , Proteínas Virais/biossíntese , Replicação Viral , eIF-2 Quinase/imunologia , eIF-2 Quinase/metabolismo , Animais , Linhagem Celular , Fator de Iniciação 2 em Eucariotos/metabolismo , Fibroblastos/virologia , Teste de Complementação Genética , Hepatócitos/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/virologia , Tropismo Viral , eIF-2 Quinase/deficiência
17.
PLoS One ; 4(3): e4772, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19274090

RESUMO

Infection of BHK cells by Sindbis virus (SV) gives rise to a profound inhibition of cellular protein synthesis, whereas translation of viral subgenomic mRNA that encodes viral structural proteins, continues for hours. To gain further knowledge on the mechanism by which this subgenomic mRNA is translated, the requirements for some initiation factors (eIFs) and for the presence of the initiator AUG were examined both in infected and in uninfected cells. To this end, BHK cells were transfected with different SV replicons or with in vitro made SV subgenomic mRNAs after inactivation of some eIFs. Specifically, eIF4G was cleaved by expression of the poliovirus 2A protease (2A(pro)) and the alpha subunit of eIF2 was inactivated by phosphorylation induced by arsenite treatment. Moreover, cellular location of these and other translation components was analyzed in BHK infected cells by confocal microscopy. Cleavage of eIF4G by poliovirus 2A(pro) does not hamper translation of subgenomic mRNA in SV infected cells, but bisection of this factor blocks subgenomic mRNA translation in uninfected cells or in cell-free systems. SV infection induces phosphorylation of eIF2alpha, a process that is increased by arsenite treatment. Under these conditions, translation of subgenomic mRNA occurs to almost the same extent as controls in the infected cells but is drastically inhibited in uninfected cells. Notably, the correct initiation site on the subgenomic mRNA is still partially recognized when the initiation codon AUG is modified to other codons only in infected cells. Finally, immunolocalization of different eIFs reveals that eIF2 alpha and eIF4G are excluded from the foci, where viral RNA replication occurs, while eIF3, eEF2 and ribosomes concentrate in these regions. These findings support the notion that canonical initiation takes place when the subgenomic mRNA is translated out of the infection context, while initiation can occur without some eIFs and even at non-AUG codons in infected cells.


Assuntos
Infecções por Alphavirus/genética , Biossíntese de Proteínas , RNA Viral/genética , Vírus Sindbis/genética , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Códon de Iniciação , Cricetinae , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 4G em Eucariotos/metabolismo , Mesocricetus , Fosforilação , Vírus de RNA , RNA Mensageiro/genética
18.
J Virol ; 82(1): 254-67, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959665

RESUMO

The E3L gene of vaccinia virus (VACV) encodes the E3 protein that in cultured cells inhibits the activation of interferon (IFN)-induced proteins, double-stranded RNA-dependent protein kinase (PKR), 2'-5'-oligoadenylate synthetase/RNase L (2-5A system) and adenosine deaminase (ADAR-1), thus helping the virus to evade host responses. Here, we have characterized the in vivo E3 functions in a murine inducible cell culture system (E3L-TetOFF) and in transgenic mice (TgE3L). Inducible E3 expression in cultured cells conferred on cells resistance to the antiviral action of IFN against different viruses, while expression of the E3L gene in TgE3L mice triggered enhanced sensitivity of the animals to pathogens. Virus infection monitored in TgE3L mice by different inoculation routes (intraperitoneal and tail scarification) showed that transgenic mice became more susceptible to VACV infection than control mice. TgE3L mice were also more susceptible to Leishmania major infection, leading to an increase in parasitemia compared to control mice. The enhanced sensitivity of TgE3L mice to VACV and L. major infections occurred together with alterations in the host immune system, as revealed by decreased T-cell responses to viral antigens in the spleen and lymph nodes and by differences in the levels of specific innate cell populations. These results demonstrate that expression of the E3L gene in transgenic mice partly reverses the resistance of the host to viral and parasitic infections and that these effects are associated with immune alterations.


Assuntos
Imunidade Inata/genética , Leishmaniose Cutânea/imunologia , Proteínas de Ligação a RNA/biossíntese , Vaccinia/imunologia , Proteínas Virais/biossíntese , Animais , Peso Corporal , Linhagem Celular , Feminino , Imunidade Inata/imunologia , Leishmania major/imunologia , Fígado/virologia , Linfonodos/imunologia , Subpopulações de Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovário/virologia , Parasitemia , Proteínas de Ligação a RNA/genética , Baço/imunologia , Baço/virologia , Linfócitos T/imunologia , Cauda/patologia , Vírus Vaccinia/imunologia , Proteínas Virais/genética
19.
Mol Biol Cell ; 18(7): 2768-77, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17507652

RESUMO

In this article we analyze the mechanisms by which the C-terminal four amino acids of inducible nitric oxide synthase (NOS2) interact with proteins that contain PDZ (PSD-95/DLG/ZO-1) domains resulting in the translocation of NOS2 to the cellular apical domain. It has been reported that human hepatic NOS2 associates to EBP50, a protein with two PDZ domains present in epithelial cells. We describe herein that NOS2 binds through its four carboxy-terminal residues to CAP70, a protein that contains four PDZ modules that is targeted to apical membranes. Interestingly, this interaction augments both the cytochrome c reductase and .NO-synthase activities of NOS2. Binding of CAP70 to NOS2 also results in an increase in the population of active NOS2 dimers. In addition, CAP70 participates in the correct subcellular targeting of NOS2 in a process that is also dependent on the acylation state of the N-terminal end of NOS2. Hence, nonpalmitoylated NOS2 is unable to progress toward the apical side of the cell despite its interaction with either EBP50 or CAP70. Likewise, if we abrogate the interaction of NOS2 with either EBP50 or CAP70 by fusing the GFP reporter to the carboxy-terminal end of NOS2 palmitoylation is not sufficient to confer an apical targeting.


Assuntos
Proteínas de Transporte/metabolismo , Polaridade Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Aminoácidos/metabolismo , Animais , Proteínas de Transporte/genética , Citosol/metabolismo , Cães , Regulação para Baixo/genética , Células Epiteliais/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isoenzimas/química , Isoenzimas/metabolismo , Fígado/enzimologia , Proteínas de Membrana , Camundongos , Óxido Nítrico Sintase Tipo II/química , Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Transfecção
20.
Virology ; 363(2): 430-6, 2007 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-17360015

RESUMO

Brefeldin A is a macrolide compound that interferes with the secretory pathway and also affects protein synthesis in mammalian cells. As a result, this antibiotic impedes the maturation of viral glycoproteins of enveloped viruses and viral genome replication in several virus species. In the present work, we show that translation of subgenomic mRNA from Sindbis virus, which in contrast to cellular translation is resistant to brefeldin A after prolonged treatment. The phosphorylation of eIF2alpha as a result of brefeldin A treatment correlates with the inhibition of cellular translation, while late viral protein synthesis is resistant to this phosphorylation. The effect of brefeldin A on Sindbis virus replication was also examined using a Sindbis virus replicon. Although brefeldin A delayed viral RNA synthesis, translation by non-replicative viral RNAs was not affected, reinforcing the idea that brefeldin A delays viral RNA replication, but does not directly affect Sindbis virus protein synthesis.


Assuntos
Infecções por Alphavirus/virologia , Brefeldina A/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Vírus Sindbis/fisiologia , Animais , Linhagem Celular/metabolismo , Linhagem Celular/virologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Fosforilação , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Replicon , Vírus Sindbis/efeitos dos fármacos , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...