Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nucleic Acids Res ; 51(20): 10950-10969, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37811880

RESUMEN

An RNA structure or modified RNA sequences can provide a platform for ribosome loading and internal translation initiation. The functional significance of internal translation has recently been highlighted by the discovery that a subset of circular RNAs (circRNAs) is internally translated. However, the molecular mechanisms underlying the internal initiation of translation in circRNAs remain unclear. Here, we identify eIF3g (a subunit of eIF3 complex) as a binding partner of eIF4A3, a core component of the exon-junction complex (EJC) that is deposited onto spliced mRNAs and plays multiple roles in the regulation of gene expression. The direct interaction between eIF4A3-eIF3g serves as a molecular linker between the eIF4A3 and eIF3 complex, thereby facilitating internal ribosomal entry. Protein synthesis from in vitro-synthesized circRNA demonstrates eIF4A3-driven internal translation, which relies on the eIF4A3-eIF3g interaction. Furthermore, our transcriptome-wide analysis shows that efficient polysomal association of endogenous circRNAs requires eIF4A3. Notably, a subset of endogenous circRNAs can express a full-length intact protein, such as ß-catenin, in an eIF4A3-dependent manner. Collectively, our results expand the understanding of the protein-coding potential of the human transcriptome, including circRNAs.


Asunto(s)
Factor 3 de Iniciación Eucariótica , Factor 4A Eucariótico de Iniciación , ARN Circular , Humanos , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Proteínas , Ribosomas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
J Biol Chem ; 299(5): 104649, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36965618

RESUMEN

The assembly of membrane-less organelles such as stress granules (SGs) is emerging as central in helping cells rapidly respond and adapt to stress. Following stress sensing, the resulting global translational shutoff leads to the condensation of stalled mRNAs and proteins into SGs. By reorganizing cytoplasmic contents, SGs can modulate RNA translation, biochemical reactions, and signaling cascades to promote survival until the stress is resolved. While mechanisms for SG disassembly are not widely understood, the resolution of SGs is important for maintaining cell viability and protein homeostasis. Mutations that lead to persistent or aberrant SGs are increasingly associated with neuropathology and a hallmark of several neurodegenerative diseases. Mutations in CLN3 are causative of juvenile neuronal ceroid lipofuscinosis, a rare neurodegenerative disease affecting children also known as Batten disease. CLN3 encodes a transmembrane lysosomal protein implicated in autophagy, endosomal trafficking, metabolism, and response to oxidative stress. Using a HeLa cell model lacking CLN3, we now show that CLN3KO is associated with an altered metabolic profile, reduced global translation, and altered stress signaling. Furthermore, loss of CLN3 function results in perturbations in SG dynamics, resulting in assembly and disassembly defects, and altered expression of the key SG nucleating factor G3BP1. With a growing interest in SG-modulating drugs for the treatment of neurodegenerative diseases, novel insights into the molecular basis of CLN3 Batten disease may reveal avenues for disease-modifying treatments for this debilitating childhood disease.


Asunto(s)
Expresión Génica , Chaperonas Moleculares , Lipofuscinosis Ceroideas Neuronales , Gránulos de Estrés , Humanos , Células HeLa , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/fisiopatología , Gránulos de Estrés/genética , Gránulos de Estrés/patología , Estrés Fisiológico/genética , Transducción de Señal/genética , Expresión Génica/genética , Línea Celular
3.
J Cell Sci ; 135(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35098996

RESUMEN

To rapidly adapt to stresses such as infections, cells have evolved several mechanisms, which include the activation of stress response pathways and the innate immune response. These stress responses result in the rapid inhibition of translation and condensation of stalled mRNAs with RNA-binding proteins and signalling components into cytoplasmic biocondensates called stress granules (SGs). Increasing evidence suggests that SGs contribute to antiviral defence, and thus viruses need to evade these responses to propagate. We previously showed that feline calicivirus (FCV) impairs SG assembly by cleaving the scaffolding protein G3BP1. We also observed that uninfected bystander cells assembled G3BP1-positive granules, suggesting a paracrine response triggered by infection. We now present evidence that virus-free supernatant generated from infected cells can induce the formation of SG-like foci, which we name paracrine granules. They are linked to antiviral activity and exhibit specific kinetics of assembly-disassembly, and protein and RNA composition that are different from canonical SGs. We propose that this paracrine induction reflects a novel cellular defence mechanism to limit viral propagation and promote stress responses in bystander cells.


Asunto(s)
Infecciones por Caliciviridae , Gránulos de Estrés , Animales , Infecciones por Caliciviridae/inmunología , Calicivirus Felino/inmunología , Gatos , Proteínas de Unión a Poli-ADP-Ribosa/inmunología , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Gránulos de Estrés/inmunología , Replicación Viral/fisiología
4.
Nature ; 561(7724): 556-560, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30232453

RESUMEN

N6-methyladenosine (m6A) modification of mRNA is emerging as an important regulator of gene expression that affects different developmental and biological processes, and altered m6A homeostasis is linked to cancer1-5. m6A modification is catalysed by METTL3 and enriched in the 3' untranslated region of a large subset of mRNAs at sites close to the stop codon5. METTL3 can promote translation but the mechanism and relevance of this process remain unknown1. Here we show that METTL3 enhances translation only when tethered to reporter mRNA at sites close to the stop codon, supporting a mechanism of mRNA looping for ribosome recycling and translational control. Electron microscopy reveals the topology of individual polyribosomes with single METTL3 foci in close proximity to 5' cap-binding proteins. We identify a direct physical and functional interaction between METTL3 and the eukaryotic translation initiation factor 3 subunit h (eIF3h). METTL3 promotes translation of a large subset of oncogenic mRNAs-including bromodomain-containing protein 4-that is also m6A-modified in human primary lung tumours. The METTL3-eIF3h interaction is required for enhanced translation, formation of densely packed polyribosomes and oncogenic transformation. METTL3 depletion inhibits tumorigenicity and sensitizes lung cancer cells to BRD4 inhibition. These findings uncover a mechanism of translation control that is based on mRNA looping and identify METTL3-eIF3h as a potential therapeutic target for patients with cancer.


Asunto(s)
Carcinogénesis , Factor 3 de Iniciación Eucariótica/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metiltransferasas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/química , ARN Mensajero/metabolismo , Animales , Línea Celular Tumoral , Ciclización , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Conformación de Ácido Nucleico , Polirribosomas/química , Polirribosomas/metabolismo , Unión Proteica , ARN Mensajero/genética
5.
Angew Chem Int Ed Engl ; 63(16): e202400422, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38380500

RESUMEN

An overreactive stress granule (SG) pathway and long-lived, stable SGs formation are thought to participate in the progress of neurodegenerative diseases (NDs). To understand if and how SGs contribute to disorders of neurotransmitter release in NDs, we examined the interaction between extracellular isolated SGs and vesicles. Amperometry shows that the vesicular content increases and dynamics of vesicle opening slow down after vesicles are treated with SGs, suggesting larger vesicles are formed. Data from transmission electron microscopy (TEM) clearly shows that a portion of large dense-core vesicles (LDCVs) with double/multiple cores appear, thus confirming that SGs induce homotypic fusion between LDCVs. This might be a protective step to help cells to survive following high oxidative stress. A hypothetical mechanism is proposed whereby enriched mRNA or protein in the shell of SGs is likely to bind intrinsically disordered protein (IDP) regions of vesicle associated membrane protein (VAMP) driving a disrupted membrane between two closely buddled vesicles to fuse with each other to form double-core vesicles. Our results show that SGs induce homotypic fusion of LDCVs, providing better understanding of how SGs intervene in pathological processes and opening a new direction to investigations of SGs involved neurodegenerative disease.


Asunto(s)
Catecolaminas , Enfermedades Neurodegenerativas , Humanos , Catecolaminas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Gránulos de Estrés , Microscopía Electrónica , Microscopía Electrónica de Transmisión
6.
J Neurosci ; 42(16): 3494-3509, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35273086

RESUMEN

Several cellular pathways contribute to neurodegenerative tauopathy-related disorders. Microglial activation, a major component of neuroinflammation, is an early pathologic hallmark that correlates with cognitive decline, while the unfolded protein response (UPR) contributes to synaptic pathology. Sleep disturbances are prevalent in tauopathies and may also contribute to disease progression. Few studies have investigated whether manipulations of sleep influence cellular pathologic and behavioral features of tauopathy. We investigated whether trazodone, a licensed antidepressant with hypnotic efficacy in dementia, can reduce disease-related cellular pathways and improve memory and sleep in male rTg4510 mice with a tauopathy-like phenotype. In a 9 week dosing regimen, trazodone decreased microglial NLRP3 inflammasome expression and phosphorylated p38 mitogen-activated protein kinase levels, which correlated with the NLRP3 inflammasome, the UPR effector ATF4, and total tau levels. Trazodone reduced theta oscillations during rapid eye movement (REM) sleep and enhanced REM sleep duration. Olfactory memory transiently improved, and memory performance correlated with REM sleep duration and theta oscillations. These findings on the effects of trazodone on the NLRP3 inflammasome, the unfolded protein response and behavioral hallmarks of dementia warrant further studies on the therapeutic value of sleep-modulating compounds for tauopathies.SIGNIFICANCE STATEMENT Dementia and associated behavioral symptoms such as memory loss and sleep disturbance are debilitating. Identifying treatments that alleviate symptoms and concurrently target cellular pathways contributing to disease progression is paramount for the patients and their caregivers. Here we show that a chronic treatment with trazodone, an antidepressant with positive effects on sleep, has beneficial effects on several cellular pathways contributing to neuroinflammation and tau pathology, in tauopathy-like rTg4510 mice. Trazodone also improved rapid eye movement (REM) sleep, the slowing of brain oscillations, and olfactory memory disturbances, which are all early symptoms observed in Alzheimer's disease. Thus, trazodone and compounds with REM sleep-promoting properties may represent a promising treatment approach to reduce the early symptoms of tauopathy and slow down disease progression.


Asunto(s)
Enfermedad de Alzheimer , Trastornos del Sueño-Vigilia , Tauopatías , Trazodona , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inflamasomas , Masculino , Trastornos de la Memoria/genética , Ratones , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Sueño/fisiología , Tauopatías/metabolismo , Trazodona/farmacología , Trazodona/uso terapéutico , Proteínas tau/metabolismo
7.
Int J Mol Sci ; 24(3)2023 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-36768868

RESUMEN

Stress granules (SGs) are stress-induced biomolecular condensates which originate primarily from inactivated RNA translation machinery and translation initiation factors. SG formation is an important defensive mechanism for cell survival, while its dysfunction has been linked to neurodegenerative diseases. However, the molecular mechanisms of SG assembly and disassembly, as well as their impacts on cellular recovery, are not fully understood. More thorough investigations into the molecular dynamics of SG pathways are required to understand the pathophysiological roles of SGs in cellular systems. Here, we characterize the SG and cytoplasmic protein turnover in neuronal progenitor cells (NPCs) under stressed and non-stressed conditions using correlative STED and NanoSIMS imaging. We incubate NPCs with isotopically labelled (15N) leucine and stress them with the ER stressor thapsigargin (TG). A correlation of STED and NanoSIMS allows the localization of individual SGs (using STED), and their protein turnover can then be extracted based on the 15N/14N ratio (using NanoSIMS). We found that TG-induced SGs, which are highly dynamic domains, recruit their constituents predominantly from the cytoplasm. Moreover, ER stress impairs the total cellular protein turnover regimen, and this impairment is not restored after the commonly proceeded stress recovery period.


Asunto(s)
Gránulos Citoplasmáticos , Enfermedades Neurodegenerativas , Humanos , Gránulos Citoplasmáticos/metabolismo , Gránulos de Estrés , Citoplasma , Enfermedades Neurodegenerativas/metabolismo , Células Madre , Estrés Fisiológico
8.
J Virol ; 95(20): e0113421, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34346771

RESUMEN

Murine norovirus (MNV) infection results in a late translation shutoff that is proposed to contribute to the attenuated and delayed innate immune response observed both in vitro and in vivo. Recently, we further demonstrated the activation of the α subunit of eukaryotic initiation factor 2 (eIF2α) kinase GCN2 during MNV infection, which has been previously linked to immunomodulation and resistance to inflammatory signaling during metabolic stress. While viral infection is usually associated with activation of double-stranded RNA (dsRNA) binding pattern recognition receptor PKR, we hypothesized that the establishment of a metabolic stress in infected cells is a proviral event, exploited by MNV to promote replication through weakening the activation of the innate immune response. In this study, we used multi-omics approaches to characterize cellular responses during MNV replication. We demonstrate the activation of pathways related to the integrated stress response, a known driver of anti-inflammatory phenotypes in macrophages. In particular, MNV infection causes an amino acid imbalance that is associated with GCN2 and ATF2 signaling. Importantly, this reprogramming lacks the features of a typical innate immune response, with the ATF/CHOP target GDF15 contributing to the lack of antiviral responses. We propose that MNV-induced metabolic stress supports the establishment of host tolerance to viral replication and propagation. IMPORTANCE During viral infection, host defenses are typically characterized by the secretion of proinflammatory autocrine and paracrine cytokines, potentiation of the interferon (IFN) response, and induction of the antiviral response via activation of JAK and Stat signaling. To avoid these and propagate, viruses have evolved strategies to evade or counteract host sensing. In this study, we demonstrate that murine norovirus controls the antiviral response by activating a metabolic stress response that activates the amino acid response and impairs inflammatory signaling. This highlights novel tools in the viral countermeasures arsenal and demonstrates the importance of the currently poorly understood metabolic reprogramming occurring during viral infections.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Macrófagos/virología , Factor de Transcripción Activador 2/metabolismo , Animales , Antivirales , Infecciones por Caliciviridae/metabolismo , Línea Celular , Factor 2 Eucariótico de Iniciación/metabolismo , Inmunidad Innata/inmunología , Inflamación/inmunología , Interferones , Macrófagos/inmunología , Ratones , Norovirus/patogenicidad , Proteínas Serina-Treonina Quinasas/metabolismo , Células RAW 264.7 , ARN Bicatenario/genética , Transducción de Señal/inmunología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
9.
PLoS Pathog ; 16(1): e1008250, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31905230

RESUMEN

Viral infections impose major stress on the host cell. In response, stress pathways can rapidly deploy defence mechanisms by shutting off the protein synthesis machinery and triggering the accumulation of mRNAs into stress granules to limit the use of energy and nutrients. Because this threatens viral gene expression, viruses need to evade these pathways to propagate. Human norovirus is responsible for gastroenteritis outbreaks worldwide. Here we examined how norovirus interacts with the eIF2α signaling axis controlling translation and stress granules. While norovirus infection represses host cell translation, our mechanistic analyses revealed that eIF2α signaling mediated by the stress kinase GCN2 is uncoupled from translational stalling. Moreover, infection results in a redistribution of the RNA-binding protein G3BP1 to replication complexes and remodelling of its interacting partners, allowing the avoidance from canonical stress granules. These results define novel strategies by which norovirus undergo efficient replication whilst avoiding the host stress response and manipulating the G3BP1 interactome.


Asunto(s)
Infecciones por Caliciviridae/virología , ADN Helicasas/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Norovirus/fisiología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Biosíntesis de Proteínas , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Animales , Infecciones por Caliciviridae/genética , Línea Celular , Gránulos Citoplasmáticos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , ARN/metabolismo , Transducción de Señal , Replicación Viral
10.
Nucleic Acids Res ; 47(18): 9789-9802, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31392996

RESUMEN

Members of the Flaviviridae family, including dengue virus (DENV) and yellow fever virus, cause serious disease in humans, whilst maternal infection with Zika virus (ZIKV) can induce microcephaly in newborns. Following infection, flaviviral RNA genomes are translated to produce the viral replication machinery but must then serve as a template for the transcription of new genomes. However, the ribosome and viral polymerase proceed in opposite directions along the RNA, risking collisions and abortive replication. Whilst generally linear, flavivirus genomes can adopt a circular conformation facilitated by long-range RNA-RNA interactions, shown to be essential for replication. Using an in vitro reconstitution approach, we demonstrate that circularization inhibits de novo translation initiation on ZIKV and DENV RNA, whilst the linear conformation is translation-competent. Our results provide a mechanism to clear the viral RNA of ribosomes in order to promote efficient replication and, therefore, define opposing roles for linear and circular conformations of the flavivirus genome.


Asunto(s)
Virus del Dengue/genética , Flavivirus/genética , Biosíntesis de Proteínas , Virus Zika/genética , Virus del Dengue/patogenicidad , Flavivirus/patogenicidad , Genoma Viral/genética , Genómica , Humanos , Recién Nacido , ARN Viral/genética , Replicación Viral/genética , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/patogenicidad , Virus Zika/patogenicidad , Infección por el Virus Zika/genética , Infección por el Virus Zika/virología
11.
Angew Chem Int Ed Engl ; 60(28): 15302-15306, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33876544

RESUMEN

Stress granules (SGs) are membrane-less organelles that assemble in the cytoplasm to organize cellular contents and promote rapid adaptation during stress. To understand how SGs contribute to physiological functions, we used electrochemical measurements to detect electroactive species in SGs. With amperometry, we discovered that reactive oxygen species (ROS) are encapsulated inside arsenite-induced SGs, and H2 O2 is the main species. The release kinetics of H2 O2 from single SGs and the number of H2 O2 molecules were quantified. The discovery that SGs contain ROS implicates them as communicators of the cellular stresses rather than a simple endpoint. This may explain how SGs regulate cellular metabolism and stress responses. This may also help better understand their cytoprotective functions in pathological conditions associated with SGs such as neurodegenerative diseases (NDs), cancers and viral infections.


Asunto(s)
Técnicas Electroquímicas , Neoplasias/diagnóstico , Especies Reactivas de Oxígeno/análisis , Humanos , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Virosis/diagnóstico , Virosis/metabolismo
12.
Nucleic Acids Res ; 46(14): e86, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-29846708

RESUMEN

We are just beginning to unravel the myriad of interactions in which non-coding RNAs participate. The intricate RNA interactome is the foundation of many biological processes, including bacterial virulence and human disease, and represents unexploited resources for the development of potential therapeutic interventions. However, identifying specific associations of a given RNA from the multitude of possible binding partners within the cell requires robust high-throughput systems for their rapid screening. Here, we present the first demonstration of functional-RNA arrays as a novel platform technology designed for the study of such interactions using immobilized, active RNAs. We have generated high-density RNA arrays by an innovative method involving surface-capture of in vitro transcribed RNAs. This approach has significant advantages over existing technologies, particularly in its versatility in regards to binding partner character. Indeed, proof-of-principle application of RNA arrays to both RNA-small molecule and RNA-RNA pairings is demonstrated, highlighting their potential as a platform technology for mapping RNA-based networks and for pharmaceutical screening. Furthermore, the simplicity of the method supports greater user-accessibility over currently available technologies. We anticipate that functional-RNA arrays will find broad utility in the expanding field of RNA characterization.


Asunto(s)
Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Pequeño no Traducido/análisis , Regiones no Traducidas 5' , Aptámeros de Nucleótidos/análisis , ARN Bacteriano/análisis
13.
J Gen Virol ; 100(11): 1501-1514, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31490115

RESUMEN

Equine hepacivirus (EHcV) (now also classified as hepacivirus A) is the closest genetic relative to hepatitis C virus (HCV) and is proposed to have diverged from HCV within the last 1000 years. The 5' untranslated regions (UTRs) of both HCV and EHcV exhibit internal ribosome entry site (IRES) activity, allowing cap-independent translational initiation, yet only the HCV 5'UTR has been systematically analysed. Here, we report a detailed structural and functional analysis of the EHcV 5'UTR. The secondary structure was determined using selective 2' hydroxyl acylation analysed by primer extension (SHAPE), revealing four stem-loops, termed SLI, SLIA, SLII and SLIII, by analogy to HCV. This guided a mutational analysis of the EHcV 5'UTR, allowing us to investigate the roles of the stem-loops in IRES function. This approach revealed that SLI was not required for EHcV IRES-mediated translation. Conversely, SLIII was essential, specifically SLIIIb, SLIIId and a GGG motif that is conserved across the Hepaciviridae. Further SHAPE analysis provided evidence that this GGG motif mediated interaction with the 40S ribosomal subunit, whilst a CUU sequence in the apical loop of SLIIIb mediated an interaction with eIF3. In addition, we showed that a microRNA122 target sequence located between SLIA and SLII mediated an enhancement of translation in the context of a subgenomic replicon. Taken together, these results highlight the conservation of hepaciviral translation mechanisms, despite divergent primary sequences.


Asunto(s)
Regiones no Traducidas 5' , Hepacivirus/genética , Sitios Internos de Entrada al Ribosoma , Animales , Línea Celular , Análisis Mutacional de ADN , Equidae/virología , Hepacivirus/crecimiento & desarrollo , Humanos , Conformación de Ácido Nucleico , Biosíntesis de Proteínas , ARN Bicatenario/genética , ARN Viral/genética , Genética Inversa
14.
RNA ; 23(11): 1712-1728, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28821580

RESUMEN

The mechanistic/mammalian target of rapamycin (mTOR) is a conserved serine/threonine kinase that integrates cellular signals from the nutrient and energy status to act, namely, on the protein synthesis machinery. While major advances have emerged regarding the regulators and effects of the mTOR signaling pathway, little is known about the regulation of mTOR gene expression. Here, we show that the human mTOR transcript can be translated in a cap-independent manner, and that its 5' untranslated region (UTR) is a highly folded RNA scaffold capable of binding directly to the 40S ribosomal subunit. We further demonstrate that mTOR is able to bypass the cap requirement for translation both in normal and hypoxic conditions. Moreover, our data reveal that the cap-independent translation of mTOR is necessary for its ability to induce cell-cycle progression into S phase. These results suggest a novel regulatory mechanism for mTOR gene expression that integrates the global protein synthesis changes induced by translational inhibitory conditions.


Asunto(s)
Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Regiones no Traducidas 5' , Animales , Hipoxia de la Célula/genética , Línea Celular , Evolución Molecular , Regulación de la Expresión Génica , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Hidrazonas/farmacología , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Biosíntesis de Proteínas , Inhibidores de la Síntesis de la Proteína/farmacología , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , Pliegue del ARN , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/genética , Tiazoles/farmacología
15.
Mol Cell Proteomics ; 16(4 suppl 1): S215-S229, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28087593

RESUMEN

Noroviruses produce viral RNAs lacking a 5' cap structure and instead use a virus-encoded viral protein genome-linked (VPg) protein covalently linked to viral RNA to interact with translation initiation factors and drive viral protein synthesis. Norovirus infection results in the induction of the innate response leading to interferon stimulated gene (ISG) transcription. However, the translation of the induced ISG mRNAs is suppressed. A SILAC-based mass spectrometry approach was employed to analyze changes to protein abundance in both whole cell and m7GTP-enriched samples to demonstrate that diminished host mRNA translation correlates with changes to the composition of the eukaryotic initiation factor complex. The suppression of host ISG translation correlates with the activity of the viral protease (NS6) and the activation of cellular caspases leading to the establishment of an apoptotic environment. These results indicate that noroviruses exploit the differences between viral VPg-dependent and cellular cap-dependent translation in order to diminish the host response to infection.


Asunto(s)
Infecciones por Caliciviridae/genética , Norovirus/metabolismo , Proteómica/métodos , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Proteínas Virales/metabolismo , Apoptosis , Infecciones por Caliciviridae/inmunología , Infecciones por Caliciviridae/virología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Marcaje Isotópico/métodos , Espectrometría de Masas/métodos , Norovirus/genética , ARN Viral/metabolismo
16.
Nucleic Acids Res ; 45(22): 13016-13028, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29069411

RESUMEN

Viral internal ribosomes entry site (IRES) elements coordinate the recruitment of the host translation machinery to direct the initiation of viral protein synthesis. Within hepatitis C virus (HCV)-like IRES elements, the sub-domain IIId(1) is crucial for recruiting the 40S ribosomal subunit. However, some HCV-like IRES elements possess an additional sub-domain, termed IIId2, whose function remains unclear. Herein, we show that IIId2 sub-domains from divergent viruses have different functions. The IIId2 sub-domain present in Seneca valley virus (SVV), a picornavirus, is dispensable for IRES activity, while the IIId2 sub-domains of two pestiviruses, classical swine fever virus (CSFV) and border disease virus (BDV), are required for 80S ribosomes assembly and IRES activity. Unlike in SVV, the deletion of IIId2 from the CSFV and BDV IRES elements impairs initiation of translation by inhibiting the assembly of 80S ribosomes. Consequently, this negatively affects the replication of CSFV and BDV. Finally, we show that the SVV IIId2 sub-domain is required for efficient viral RNA synthesis and growth of SVV, but not for IRES function. This study sheds light on the molecular evolution of viruses by clearly demonstrating that conserved RNA structures, within distantly related RNA viruses, have acquired different roles in the virus life cycles.


Asunto(s)
Sitios Internos de Entrada al Ribosoma/genética , Pestivirus/genética , Picornaviridae/genética , ARN Viral/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Virus de la Enfermedad de la Frontera/genética , Virus de la Enfermedad de la Frontera/fisiología , Línea Celular , Virus de la Fiebre Porcina Clásica/genética , Virus de la Fiebre Porcina Clásica/fisiología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Conformación de Ácido Nucleico , Pestivirus/fisiología , Picornaviridae/fisiología , ARN Viral/química , ARN Viral/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Porcinos
17.
J Gen Virol ; 98(9): 2207-2214, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28869001

RESUMEN

The role of m6A methylation of RNA has remained elusive for decades, but recent technological advances are now allowing the mapping of the m6A methylation landscape at nucleotide level. This has spurred an explosion in our understanding of the role played by RNA epigenetics in RNA biology. m6A modifications have been tied to almost every aspect of the mRNA life cycle and it is now clear that RNA virus genomes are subject to m6A methylation. These modifications play various roles in the viral replication cycle. This review will summarize recent breakthroughs concerning m6A RNA modification and their implications for cellular and viral RNAs.


Asunto(s)
Infecciones por Virus ARN/virología , Virus ARN/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Animales , Humanos , Metilación , Virus ARN/genética , ARN Mensajero/genética , ARN Viral/genética
18.
J Virol ; 90(14): 6489-6501, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27147742

RESUMEN

UNLABELLED: In response to stress such as virus infection, cells can stall translation by storing mRNAs away in cellular compartments called stress granules (SGs). This defense mechanism favors cell survival by limiting the use of energy and nutrients until the stress is resolved. In some cases it may also block viral propagation as viruses are dependent on the host cell resources to produce viral proteins. Human norovirus is a member of the Caliciviridae family responsible for gastroenteritis outbreaks worldwide. Previous studies on caliciviruses have identified mechanisms by which they can usurp the host translational machinery, using the viral protein genome-linked VPg, or regulate host protein synthesis through the mitogen-activated protein kinase (MAPK) pathway. Here, we examined the effect of feline calicivirus (FCV) infection on SG accumulation. We show that FCV infection impairs the assembly of SGs despite an increased phosphorylation of eukaryotic initiation factor eIF2α, a hallmark of stress pathway activation. Furthermore, SGs did not accumulate in FCV-infected cells that were stressed with arsenite or hydrogen peroxide. FCV infection resulted in the cleavage of the SG-nucleating protein Ras-GTPase activating SH3 domain-binding protein (G3BP1), which is mediated by the viral 3C-like proteinase NS6(Pro) Using mutational analysis, we identified the FCV-induced cleavage site within G3BP1, which differs from the poliovirus 3C proteinase cleavage site previously identified. Finally, we showed that NS6(Pro)-mediated G3BP1 cleavage impairs SG assembly. In contrast, murine norovirus (MNV) infection did not impact arsenite-induced SG assembly or G3BP1 integrity, suggesting that related caliciviruses have distinct effects on the stress response pathway. IMPORTANCE: Human noroviruses are a major cause of viral gastroenteritis, and it is important to understand how they interact with the infected host cell. Feline calicivirus (FCV) and murine norovirus (MNV) are used as models to understand norovirus biology. Recent studies have suggested that the assembly of stress granules is central in orchestrating stress and antiviral responses to restrict viral replication. Overall, our study provides the first insight on how caliciviruses impair stress granule assembly by targeting the nucleating factor G3BP1 via the viral proteinase NS6(Pro) This work provides new insights into host-pathogen interactions that regulate stress pathways during FCV infection.


Asunto(s)
Infecciones por Caliciviridae/virología , Calicivirus Felino/patogenicidad , Proteínas Portadoras/metabolismo , Gránulos Citoplasmáticos/metabolismo , Interacciones Huésped-Patógeno , Replicación Viral , Proteasas Virales 3C , Animales , Infecciones por Caliciviridae/metabolismo , Infecciones por Caliciviridae/patología , Proteínas Portadoras/genética , Gatos , Cisteína Endopeptidasas/metabolismo , Gránulos Citoplasmáticos/virología , ADN Helicasas , Factor 2 Eucariótico de Iniciación/metabolismo , Células HeLa , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Proteínas Virales/metabolismo
19.
J Biol Chem ; 290(8): 4748-4758, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25561727

RESUMEN

Protein synthesis is a tightly controlled process responding to several stimuli, including viral infection. As obligate intracellular parasites, viruses depend on the translation machinery of the host and can manipulate it by affecting the availability and function of specific eukaryotic initiation factors (eIFs). Human norovirus is a member of the Caliciviridae family and is responsible for gastroenteritis outbreaks. Previous studies on feline calicivirus and murine norovirus 1 (MNV1) demonstrated that the viral protein, genome-linked (VPg), acts to direct translation by hijacking the host protein synthesis machinery. Here we report that MNV1 infection modulates the MAPK pathway to activate eIF4E phosphorylation. Our results show that the activation of p38 and Mnk during MNV1 infection is important for MNV1 replication. Furthermore, phosphorylated eIF4E relocates to the polysomes, and this contributes to changes in the translational state of specific host mRNAs. We propose that global translational control of the host by eIF4E phosphorylation is a key component of the host-pathogen interaction.


Asunto(s)
Infecciones por Caliciviridae/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Interacciones Huésped-Patógeno , Norovirus/fisiología , Biosíntesis de Proteínas , Proteínas Virales/biosíntesis , Replicación Viral/fisiología , Animales , Infecciones por Caliciviridae/genética , Gatos , Línea Celular , Factor 4E Eucariótico de Iniciación/genética , Humanos , Ratones , Fosforilación/genética , Polirribosomas/genética , Polirribosomas/metabolismo , Transporte de Proteínas/genética , Proteínas Virales/genética
20.
RNA ; 20(11): 1803-14, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25246653

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus, the etiological agent of Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL). One of the key viral proteins that contributes to tumorigenesis is vFLIP, a viral homolog of the FLICE inhibitory protein. This KSHV protein interacts with the NFκB pathway to trigger the expression of antiapoptotic and proinflammatory genes and ultimately leads to tumor formation. The expression of vFLIP is regulated at the translational level by an internal ribosomal entry site (IRES) element. However, the precise mechanism by which ribosomes are recruited internally and the exact location of the IRES has remained elusive. Here we show that a 252-nt fragment directly upstream of vFLIP, within a coding region, directs translation. We have established its RNA structure and demonstrate that IRES activity requires the presence of eIF4A and an intact eIF4G. Furthermore, and unusually for an IRES, eIF4E is part of the complex assembled onto the vFLIP IRES to direct translation. These molecular interactions define a new paradigm for IRES-mediated translation.


Asunto(s)
Herpesvirus Humano 8/genética , ARN Viral/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Sitios de Unión , Línea Celular Tumoral , Regulación Viral de la Expresión Génica , Células HEK293 , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , ARN Viral/genética , Ribosomas/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA