Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 83(3): 481-495, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36334591

RESUMO

Viral reproduction is contingent on viral protein synthesis that relies on the host ribosomes. As such, viruses have evolved remarkable strategies to hijack the host translational apparatus in order to favor viral protein production and to interfere with cellular innate defenses. Here, we describe the approaches viruses use to exploit the translation machinery, focusing on commonalities across diverse viral families, and discuss the functional relevance of this process. We illustrate the complementary strategies host cells utilize to block viral protein production and consider how cells ensure an efficient antiviral response that relies on translation during this tug of war over the ribosome. Finally, we highlight potential roles mRNA modifications and ribosome quality control play in translational regulation and innate immunity. We address these topics in the context of the COVID-19 pandemic and focus on the gaps in our current knowledge of these mechanisms, specifically in viruses with pandemic potential.


Assuntos
COVID-19 , Biossíntese de Proteínas , Viroses , Vírus , Humanos , COVID-19/genética , COVID-19/imunologia , Pandemias , Biossíntese de Proteínas/genética , Biossíntese de Proteínas/imunologia , RNA Viral/genética , RNA Viral/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Viroses/genética , Viroses/imunologia , Vírus/genética , Vírus/imunologia , Ribossomos/genética , Ribossomos/imunologia , Ribossomos/virologia
2.
Viruses ; 14(2)2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35215780

RESUMO

Viruses are obligate intracellular parasites that depend on the host's protein synthesis machinery for translating their mRNAs. The viral mRNA (vRNA) competes with the host mRNA to recruit the translational machinery, including ribosomes, tRNAs, and the limited eukaryotic translation initiation factor (eIFs) pool. Many viruses utilize non-canonical strategies such as targeting host eIFs and RNA elements known as internal ribosome entry sites (IRESs) to reprogram cellular gene expression, ensuring preferential translation of vRNAs. In this review, we discuss vRNA IRES-mediated translation initiation, highlighting the role of RNA-binding proteins (RBPs), other than the canonical translation initiation factors, in regulating their activity.


Assuntos
Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Viral/genética , Proteínas de Ligação a RNA/metabolismo , Viroses/metabolismo , Vírus/genética , Animais , Humanos , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Ribossomos/genética , Ribossomos/metabolismo , Ribossomos/virologia , Viroses/genética , Viroses/virologia , Vírus/metabolismo
3.
RNA ; 27(9): 1025-1045, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34127534

RESUMO

Viruses rely on the host translation machinery to synthesize their own proteins. Consequently, they have evolved varied mechanisms to co-opt host translation for their survival. SARS-CoV-2 relies on a nonstructural protein, Nsp1, for shutting down host translation. However, it is currently unknown how viral proteins and host factors critical for viral replication can escape a global shutdown of host translation. Here, using a novel FACS-based assay called MeTAFlow, we report a dose-dependent reduction in both nascent protein synthesis and mRNA abundance in cells expressing Nsp1. We perform RNA-seq and matched ribosome profiling experiments to identify gene-specific changes both at the mRNA expression and translation levels. We discover that a functionally coherent subset of human genes is preferentially translated in the context of Nsp1 expression. These genes include the translation machinery components, RNA binding proteins, and others important for viral pathogenicity. Importantly, we uncovered a remarkable enrichment of 5' terminal oligo-pyrimidine (TOP) tracts among preferentially translated genes. Using reporter assays, we validated that 5' UTRs from TOP transcripts can drive preferential expression in the presence of Nsp1. Finally, we found that LARP1, a key effector protein in the mTOR pathway, may contribute to preferential translation of TOP transcripts in response to Nsp1 expression. Collectively, our study suggests fine-tuning of host gene expression and translation by Nsp1 despite its global repressive effect on host protein synthesis.


Assuntos
Interações Hospedeiro-Patógeno/genética , Biossíntese de Proteínas , Proteínas/química , Proteínas/genética , Proteínas não Estruturais Virais/genética , Regiões 5' não Traduzidas , Autoantígenos/genética , Autoantígenos/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Dobramento de Proteína , Pirimidinas , RNA Mensageiro/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Ribossomos/genética , Ribossomos/virologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteínas não Estruturais Virais/metabolismo , Antígeno SS-B
4.
Commun Biol ; 4(1): 715, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112887

RESUMO

While SARS-CoV-2 is causing modern human history's most serious health crisis and upending our way of life, clinical and basic research on the virus is advancing rapidly, leading to fascinating discoveries. Two studies have revealed how the viral virulence factor, nonstructural protein 1 (Nsp1), binds human ribosomes to inhibit host cell translation. Here, we examine the main conclusions on the molecular activity of Nsp1 and its role in suppressing innate immune responses. We discuss different scenarios potentially explaining how the viral RNA can bypass its own translation blockage and speculate on the suitability of Nsp1 as a therapeutic target.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Ribossomos/virologia , SARS-CoV-2/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Regiões 5' não Traduzidas , Regulação Viral da Expressão Gênica , Humanos , Imunidade Inata , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Ribossomos/metabolismo , SARS-CoV-2/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
5.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33806254

RESUMO

The global COVID-19 pandemic caused by SARS-CoV-2 has resulted in over 2.2 million deaths. Disease outcomes range from asymptomatic to severe with, so far, minimal genotypic change to the virus so understanding the host response is paramount. Transcriptomics has become incredibly important in understanding host-pathogen interactions; however, post-transcriptional regulation plays an important role in infection and immunity through translation and mRNA stability, allowing tight control over potent host responses by both the host and the invading virus. Here, we apply ribosome profiling to assess post-transcriptional regulation of host genes during SARS-CoV-2 infection of a human lung epithelial cell line (Calu-3). We have identified numerous transcription factors (JUN, ZBTB20, ATF3, HIVEP2 and EGR1) as well as select antiviral cytokine genes, namely IFNB1, IFNL1,2 and 3, IL-6 and CCL5, that are restricted at the post-transcriptional level by SARS-CoV-2 infection and discuss the impact this would have on the host response to infection. This early phase restriction of antiviral transcripts in the lungs may allow high viral load and consequent immune dysregulation typically seen in SARS-CoV-2 infection.


Assuntos
Citocinas/genética , Processamento Pós-Transcricional do RNA , Ribossomos/metabolismo , Ribossomos/virologia , SARS-CoV-2/imunologia , Fatores de Transcrição/genética , Animais , Antivirais/antagonistas & inibidores , Linhagem Celular Tumoral , Chlorocebus aethiops , Biologia Computacional , Citocinas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata/genética , Pulmão/imunologia , Pulmão/virologia , RNA Mensageiro/metabolismo , RNA-Seq , Ribossomos/genética , SARS-CoV-2/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma , Células Vero
6.
Biochemistry ; 59(46): 4429-4438, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33166472

RESUMO

Minor-groove base triples formed between stem 1 and loop 2 of the simian retrovirus type 1 (SRV-1) mRNA frameshifting pseudoknot are essential in stimulating -1 ribosomal frameshifting. How tertiary base triple formation affects the local stabilities of secondary structures (stem 1 and stem 2) and thus ribosomal frameshifting efficiency is not well understood. We made a short peptide nucleic acid (PNA) that is expected to invade stem 1 of the SRV-1 pseudoknot by PNA-RNA duplex formation to mimic the stem 1 unwinding process by a translating ribosome. In addition, we used a PNA for invading stem 2 in the SRV-1 pseudoknot. Our nondenaturing polyacrylamide gel electrophoresis data for the binding of PNA to the SRV-1 pseudoknot and mutants reveal that mutations in loop 2 disrupting base triple formation between loop 2 and stem 1 in the SRV-1 pseudoknot result in enhanced invasion by both PNAs. Our data suggest that tertiary stem 1-loop 2 base triple interactions in the SRV-1 pseudoknot can stabilize both of the secondary structural components, stem 1 and stem 2. Stem 2 stability is thus coupled to the structural stability of stem 1-loop 2 base triples, mediated through a long-range effect. The apparent dissociation constants of both PNAs are positively correlated with the pseudoknot mechanical stabilities and frameshifting efficiencies. The relatively simple PNA local invasion experiment may be used to characterize the energetic contribution of tertiary interactions and ligand binding in many other RNA and DNA structures.


Assuntos
Mudança da Fase de Leitura do Gene Ribossômico , Vírus dos Macacos de Mason-Pfizer/genética , RNA Viral/química , Eletroforese em Gel de Poliacrilamida Nativa , Conformação de Ácido Nucleico , Ácidos Nucleicos Peptídicos/química , Ácidos Nucleicos Peptídicos/genética , Ribossomos/genética , Ribossomos/virologia
7.
J Proteome Res ; 19(11): 4275-4290, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-32686937

RESUMO

SARS-CoV-2 (COVID-19) has infected millions of people worldwide, with lethality in hundreds of thousands. The rapid publication of information, both regarding the clinical course and the viral biology, has yielded incredible knowledge of the virus. In this review, we address the insights gained for the SARS-CoV-2 proteome, which we have integrated into the Viral Integrated Structural Evolution Dynamic Database, a publicly available resource. Integrating evolutionary, structural, and interaction data with human proteins, we present how the SARS-CoV-2 proteome interacts with human disorders and risk factors ranging from cytokine storm, hyperferritinemic septic, coagulopathic, cardiac, immune, and rare disease-based genetics. The most noteworthy human genetic potential of SARS-CoV-2 is that of the nucleocapsid protein, where it is known to contribute to the inhibition of the biological process known as nonsense-mediated decay. This inhibition has the potential to not only regulate about 10% of all biological transcripts through altered ribosomal biology but also associate with viral-induced genetics, where suppressed human variants are activated to drive dominant, negative outcomes within cells. As we understand more of the dynamic and complex biological pathways that the proteome of SARS-CoV-2 utilizes for entry into cells, for replication, and for release from human cells, we can understand more risk factors for severe/lethal outcomes in patients and novel pharmaceutical interventions that may mitigate future pandemics.


Assuntos
Betacoronavirus , Infecções por Coronavirus , Interações Hospedeiro-Patógeno , Pandemias , Pneumonia Viral , Proteoma , Ribossomos , COVID-19 , Infecções por Coronavirus/genética , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Humanos , Pneumonia Viral/genética , Pneumonia Viral/metabolismo , Pneumonia Viral/virologia , Proteoma/genética , Proteoma/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Ribossomos/virologia , SARS-CoV-2 , Transcriptoma , Proteínas Virais
8.
J Biol Chem ; 294(42): 15386-15394, 2019 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-31455634

RESUMO

During enteroviral infections, the canonical translation factor eukaryotic translation initiation factor 4 γ I (eIF4GI) is cleaved by viral protease 2A. The resulting C-terminal fragment is recruited by the viral internal ribosome entry site (IRES) for efficient translation of the viral RNA. However, the 2A protease is not present in the viral capsid and is synthesized only after the initial round of translation. This presents the conundrum of how the initial round of translation occurs in the absence of the C-terminal eIF4GI fragment. Interestingly, the host protein DAP5 (also known as p97, eIF4GIII, and eIF4G2), an isoform of eIF4GI, closely resembles the eIF4GI C-terminal fragment produced after 2A protease-mediated cleavage. Using the Coxsackievirus B3 (CVB3) IRES as a model system, here we demonstrate that DAP5, but not the full-length eIF4GI, is required for CVB3 IRES activity for translation of input viral RNA. Additionally, we show that DAP5 is specifically required by type I IRES but not by type II or type III IRES, in which cleavage of eIF4GI has not been observed. We observed that both DAP5 and C-terminal eIF4GI interact with CVB3 IRES in the same region, but DAP5 exhibits a lower affinity for CVB3 IRES compared with the C-terminal eIF4GI fragment. It appears that DAP5 is required for the initial round of viral RNA translation by sustaining a basal level of CVB3 IRES activity. This activity leads to expression of 2A protease and consequent robust CVB3 IRES-mediated translation by the C-terminal eIF4GI fragment.


Assuntos
Infecções por Coxsackievirus/metabolismo , Enterovirus Humano B/genética , Fator de Iniciação Eucariótico 4G/metabolismo , RNA Viral/genética , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/metabolismo , Fator de Iniciação Eucariótico 4G/genética , Regulação Viral da Expressão Gênica , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Sítios Internos de Entrada Ribossomal , Biossíntese de Proteínas , RNA Viral/metabolismo , Ribossomos/metabolismo , Ribossomos/virologia
9.
Cells ; 8(5)2019 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-31137833

RESUMO

Ribosomal proteins (RPs), in conjunction with rRNA, are major components of ribosomes involved in the cellular process of protein biosynthesis, known as "translation". The viruses, as the small infectious pathogens with limited genomes, must recruit a variety of host factors to survive and propagate, including RPs. At present, more and more information is available on the functional relationship between RPs and virus infection. This review focuses on advancements in my own understanding of critical roles of RPs in the life cycle of viruses. Various RPs interact with viral mRNA and proteins to participate in viral protein biosynthesis and regulate the replication and infection of virus in host cells. Most interactions are essential for viral translation and replication, which promote viral infection and accumulation, whereas the minority represents the defense signaling of host cells by activating immune pathway against virus. RPs provide a new platform for antiviral therapy development, however, at present, antiviral therapeutics with RPs involving in virus infection as targets is limited, and exploring antiviral strategy based on RPs will be the guides for further study.


Assuntos
Antivirais/metabolismo , RNA Viral/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/virologia , Viroses/metabolismo , Vírus/metabolismo , Mudança da Fase de Leitura do Gene Ribossômico , Interações Hospedeiro-Patógeno , Humanos , Fosforilação , RNA Mensageiro/metabolismo , Ribossomos/metabolismo , Replicação Viral
10.
Adv Virus Res ; 103: 135-166, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30635075

RESUMO

The discovery of giant viruses revealed a new level of complexity in the virosphere, raising important questions about the diversity, ecology, and evolution of these viruses. The family Mimiviridae was the first group of amoebal giant viruses to be discovered (by Bernard La Scola and Didier Raoult team), containing viruses with structural and genetic features that challenged many concepts of classic virology. The tupanviruses are among the newest members of this family and exhibit structural, biological, and genetic features never previously observed in other giant viruses. The complexity of these viruses has put us one step forward toward the comprehension of giant virus biology and evolution, but also has raised important questions that still need to be addressed. In this chapter, we tell the history behind the discovery of one of the most complex viruses isolated to date, highlighting the unique features exhibited by tupanviruses, and discuss how these giant viruses have contributed to redefining limits for the virosphere.


Assuntos
Especificidade de Hospedeiro , Mimiviridae/fisiologia , Biossíntese de Proteínas , Proteínas Virais/genética , Amoeba/virologia , Genoma Viral , Vírus Gigantes/fisiologia , Interações Hospedeiro-Patógeno , Mimiviridae/isolamento & purificação , Ribossomos/genética , Ribossomos/virologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia
11.
Nucleic Acids Res ; 47(1): 432-449, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30395302

RESUMO

The 2'-5'-oligoadenylate synthetase (OAS)/RNase L pathway is an innate immune system that protects hosts against pathogenic viruses and bacteria through cleavage of exogenous single-stranded RNA; however, this system's selective targeting mechanism remains unclear. Here, we identified an mRNA quality control factor Dom34 as a novel restriction factor for a positive-sense single-stranded RNA virus. Downregulation of Dom34 and RNase L increases viral replication, as well as half-life of the viral RNA. Dom34 directly binds RNase L to form a surveillance complex to recognize and eliminate the exogenous RNA in a manner dependent on translation. Interestingly, the feature detected by the surveillance complex is not the specific sequence of the viral RNA but the 'exogenous nature' of the RNA. We propose the following model for the selective targeting of exogenous RNA; OAS3 activated by the exogenous RNA releases 2'-5'-oligoadenylates (2-5A), which in turn converts latent RNase L to an active dimer. This accelerates formation of the Dom34-RNase L surveillance complex, and its selective localization to the ribosome on the exogenous RNA, thereby promoting degradation of the RNA. Our findings reveal that the selective targeting of exogenous RNA in antiviral defense occurs via a mechanism similar to that in the degradation of aberrant transcripts in RNA quality control.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Endonucleases/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais/genética , Viroses/genética , Vírus/genética , Nucleotídeos de Adenina/genética , Nucleotídeos de Adenina/metabolismo , Endonucleases/genética , Endorribonucleases/genética , Regulação Viral da Expressão Gênica , Humanos , Proteínas Nucleares/genética , Oligorribonucleotídeos/genética , Oligorribonucleotídeos/metabolismo , Estabilidade de RNA/genética , RNA de Cadeia Dupla/genética , RNA Viral/genética , Ribossomos/genética , Ribossomos/virologia , Viroses/virologia , Replicação Viral/genética , Vírus/patogenicidade
12.
IUBMB Life ; 70(1): 41-49, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29281185

RESUMO

Hepatitis C virus (HCV) has infected over 170 million people world-wide. This infection causes severe liver damage that can progress to hepatocellular carcinoma leading to death of the infected patients. Development of a cell culture model system for the study of HCV infection in the recent past has helped the researchers world-wide to understand the biology of this virus. Studies over the past decade have revealed the tricks played by the virus to sustain itself, for as long as 40 years, in the host setup without being eliminated by the immune system. Today we understand that the host organelles and different cellular proteins are affected during HCV infection. This cytoplasmic virus has all the cellular organelles at its disposal to successfully replicate, from ribosomes and intracellular membranous structures to the nucleus. It modulates these organelles at both the structural and the functional levels. The vast knowledge about the viral genome and viral proteins has also helped in the development of drugs against the virus. Despite the achieved success rate to cure the infected patients, we struggle to eliminate the cases of recurrence and the non-responders. Such cases might emerge owing to the property of the viral genome to accumulate mutations during its succeeding replication cycles which favours its survival. The current situation calls an urgent need for alternate therapeutic strategies to counter this major problem of human health. © 2017 IUBMB Life, 70(1):41-49, 2018.


Assuntos
Carcinoma Hepatocelular/virologia , Hepacivirus/patogenicidade , Hepatite C Crônica/virologia , Hepatócitos/virologia , Evasão da Resposta Imune , Neoplasias Hepáticas/virologia , Antivirais/uso terapêutico , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/prevenção & controle , Núcleo Celular/imunologia , Núcleo Celular/virologia , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Regulação da Expressão Gênica , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepatite C Crônica/complicações , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/imunologia , Hepatócitos/imunologia , Humanos , Gotículas Lipídicas/imunologia , Gotículas Lipídicas/virologia , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/imunologia , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/prevenção & controle , RNA Viral/biossíntese , RNA Viral/genética , Ribossomos/imunologia , Ribossomos/virologia , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/imunologia , Replicação Viral/efeitos dos fármacos
13.
J Virol ; 90(12): 5538-5540, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27053555

RESUMO

To replicate, all viruses depend entirely on the enslavement of host cell ribosomes for their own advantage. To this end, viruses have evolved a multitude of translational strategies to usurp the ribosome. RNA-based structures known as internal ribosome entry sites (IRESs) are among the most notable mechanisms employed by viruses to seize host ribosomes. In this article, we spotlight the intergenic region IRES from the Dicistroviridae family of viruses and its importance as a model for IRES-dependent translation and in understanding fundamental properties of translation.


Assuntos
Dicistroviridae/genética , Dicistroviridae/fisiologia , Genoma Viral , Sítios Internos de Entrada Ribossomal , Ribossomos/genética , Animais , Interações Hospedeiro-Patógeno , Modelos Moleculares , Biossíntese de Proteínas , RNA Viral/genética , Ribossomos/metabolismo , Ribossomos/virologia
14.
Assay Drug Dev Technol ; 11(6): 355-66, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23906347

RESUMO

The use of small molecules to modulate cellular processes is a powerful approach to investigate gene function as a complement to genetic approaches. The discovery and characterization of compounds that modulate translation initiation, the rate-limiting step of protein synthesis, is important both to provide tool compounds to explore this fundamental biological process and to further evaluate protein synthesis as a therapeutic target. While most messenger ribonucleic acids (mRNAs) recruit ribosomes via their 5' cap, some viral and cellular mRNAs initiate protein synthesis via an alternative "cap-independent" mechanism utilizing internal ribosome entry sites (IRES) elements, which are complex mRNA secondary structures, localized within the 5' nontranslated region of the mRNA upstream of the AUG start codon. This report describes the design of a functional, high throughput screen of small molecules miniaturized into a 1,536-well format and performed using the luciferase reporter gene under control of the viral Cardiovirus encephalomyocarditis virus (EMCV) IRES element to identify nontoxic compounds modulating translation initiated from the EMCV IRES. One activating compound, validated in a dose response manner, has previously been shown to bind the glucocorticoid receptor (GR). Subsequent testing of additional GR modulators further supported this as the possible mechanism of action. Detailed characterization of this compound activity supported the notion that this was due to an effect at the level of translation.


Assuntos
Vírus da Encefalomiocardite/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Receptores de Glucocorticoides/efeitos dos fármacos , Ribossomos/virologia , Internalização do Vírus/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Desenho de Fármacos , Vírus da Encefalomiocardite/fisiologia , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Receptores de Glucocorticoides/fisiologia
15.
Mol Cell Biol ; 33(5): 1016-26, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23275440

RESUMO

During viral infection or cellular stress, cap-dependent translation is shut down. Proteins that are synthesized under these conditions use alternative mechanisms to initiate translation. This study demonstrates that at least two alternative translation initiation routes, internal ribosome entry site (IRES) initiation and ribosome shunting, rely on ribosomal protein S25 (RPS25). This suggests that they share a mechanism for initiation that is not employed by cap-dependent translation, since cap-dependent translation is not affected by the loss of RPS25. Furthermore, we demonstrate that viruses that utilize an IRES or a ribosome shunt, such as hepatitis C virus, poliovirus, or adenovirus, have impaired amplification in cells depleted of RPS25. In contrast, viral amplification of a virus that relies solely on cap-dependent translation, herpes simplex virus, is not hindered. We present a model that explains how RPS25 can be a nexus for multiple alternative translation initiation pathways.


Assuntos
Adenoviridae/fisiologia , Hepacivirus/fisiologia , Interações Hospedeiro-Patógeno , Poliovirus/fisiologia , Proteínas Ribossômicas/metabolismo , Ribossomos/virologia , Infecções por Adenoviridae/genética , Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/virologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Células HeLa , Hepatite C/genética , Hepatite C/metabolismo , Hepatite C/virologia , Humanos , Poliomielite/genética , Poliomielite/metabolismo , Poliomielite/virologia , Biossíntese de Proteínas , Proteínas Ribossômicas/genética , Ribossomos/metabolismo , Replicação Viral
16.
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
17.
Biol Chem ; 393(10): 1079-88, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23091275

RESUMO

ATP binding cassette proteins are a large and diverse family of molecular machines and include transmembrane transporter, chromosome maintenance and DNA repair proteins, and translation factors. However, the function of the ABCE1, the only member of subfamily E of ABC proteins, remained mysterious for over a decade, even though it is perhaps the most conserved ABC protein in eukaryotes and archaea. Recent results have now identified ABCE1 as the ribosome-recycling factor of eukaryotes and archaea. Thus, two iron-sulfur clusters - the hallmark feature of ABCE1 - help catalyze an integral step of the translational cycle at the core of the protein synthesis machinery.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Biossíntese de Proteínas , Transportadores de Cassetes de Ligação de ATP/química , Regulação Alostérica , Animais , Imunidade Inata , Ribossomos/genética , Ribossomos/imunologia , Ribossomos/metabolismo , Ribossomos/virologia , Especificidade da Espécie
18.
Arterioscler Thromb Vasc Biol ; 32(4): 997-1004, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22328780

RESUMO

OBJECTIVE: Translation initiation of eukaryotic mRNAs typically occurs by cap-dependent ribosome scanning mechanism. However, certain mRNAs are translated by ribosome assembly at internal ribosome entry sites (IRESs). Whether IRES-mediated translation occurs in stressed primary human endothelial cells (ECs) is unknown. METHODS AND RESULTS: We performed microarray analysis of polyribosomal mRNA from ECs to identify IRES-containing mRNAs. Cap-dependent translation was disabled by poliovirus (PV) infection and confirmed by loss of polysome peaks, detection of eukaryotic initiation factor (eIF) 4G cleavage, and decreased protein synthesis. We found that 87.4% of mRNAs were dissociated from polysomes in virus-infected ECs. Twelve percent of mRNAs remained associated with polysomes, and 0.6% were enriched ≥2-fold in polysome fractions from infected ECs. Quantitative reverse transcription-polymerase chain reaction confirmed the microarray findings for 31 selected mRNAs. We found that enriched polysome associations of programmed cell death 8 (PDCD8) and JunB mRNA resulted in increased protein expression in PV-infected ECs. The presence of IRESs in the 5' untranslated region of PDCD8 mRNA, but not of JunB mRNA, was confirmed by dicistronic analysis. CONCLUSIONS: We show that microarray profiling of polyribosomal mRNA transcripts from PV-infected ECs successfully identifies mRNAs whose translation is preserved in the face of stress-induced, near complete cessation of cap-dependent initiation. Nevertheless, internal ribosome entry is not the only mechanism responsible for this privileged translation.


Assuntos
Fator de Indução de Apoptose/biossíntese , Células Endoteliais/virologia , Poliovirus/patogenicidade , Proteínas Proto-Oncogênicas c-jun/biossíntese , RNA Mensageiro/metabolismo , Ribossomos/virologia , Regiões 5' não Traduzidas , Fator de Indução de Apoptose/genética , Linhagem Celular , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica/métodos , Genes Reporter , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-jun/genética , Capuzes de RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribossomos/metabolismo , Transfecção
19.
Gene Ther ; 19(9): 947-55, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22278412

RESUMO

The treatment of axonal disorders, such as diseases associated with axonal injury and degeneration, is limited by the inability to directly target therapeutic protein expression to injured axons. Current gene therapy approaches rely on infection and transcription of viral genes in the cell body. Here, we describe an approach to target gene expression selectively to axons. Using a genetically engineered mouse containing epitope-labeled ribosomes, we find that neurons in adult animals contain ribosomes in distal axons. To use axonal ribosomes to alter local protein expression, we utilized a Sindbis virus containing an RNA genome that has been modified so that it can be directly used as a template for translation. Selective application of this virus to axons leads to local translation of heterologous proteins. Furthermore, we demonstrate that selective axonal protein expression can be used to modify axonal signaling in cultured neurons, enabling axons to grow over inhibitory substrates typically encountered following axonal injury. We also show that this viral approach also can be used to achieve heterologous expression in axons of living animals, indicating that this approach can be used to alter the axonal proteome in vivo. Together, these data identify a novel strategy to manipulate protein expression in axons, and provides a novel approach for using gene therapies for disorders of axonal function.


Assuntos
Axônios/fisiologia , Marcação de Genes/métodos , Vetores Genéticos , Sindbis virus/genética , Adenilil Ciclases/genética , Animais , Axônios/metabolismo , Camundongos , Regeneração Nervosa , Ribossomos/virologia , Medula Espinal
20.
Virology ; 425(1): 40-52, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22280896

RESUMO

Picornavirus infection can lead to disruption of nuclear pore traffic, shut-off of cell translation machinery, and cleavage of proteins involved in cellular signal transduction and the innate response to infection. Here, we demonstrated that the FMDV 3C(pro) induced the cleavage of nuclear RNA-binding protein Sam68 C-terminus containing the nuclear localization sequence (NLS). Consequently, it stimulated the redistribution of Sam68 to the cytoplasm. The siRNA knockdown of Sam68 resulted in a 1000-fold reduction in viral titers, which prompted us to study the effect of Sam68 on FMDV post-entry events. Interestingly, Sam68 interacts with the internal ribosomal entry site within the 5' non-translated region of the FMDV genome, and Sam68 knockdown decreased FMDV IRES-driven activity in vitro suggesting that it could modulate translation of the viral genome. The results uncover a novel role for Sam68 in the context of picornaviruses and the proteolysis of a new cellular target of the FMDV 3C(pro).


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cisteína Endopeptidases/metabolismo , Citoplasma/metabolismo , Vírus da Febre Aftosa/patogenicidade , Rim/virologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Vírus da Febre Aftosa/enzimologia , Rim/citologia , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Ribossomos/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA