Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 83(14): 2559-2577.e8, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37421942

RESUMEN

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remodels the endoplasmic reticulum (ER) to form replication organelles, leading to ER stress and unfolded protein response (UPR). However, the role of specific UPR pathways in infection remains unclear. Here, we found that SARS-CoV-2 infection causes marginal activation of signaling sensor IRE1α leading to its phosphorylation, clustering in the form of dense ER-membrane rearrangements with embedded membrane openings, and XBP1 splicing. By investigating the factors regulated by IRE1α-XBP1 during SARS-CoV-2 infection, we identified stress-activated kinase NUAK2 as a novel host-dependency factor for SARS-CoV-2, HCoV-229E, and MERS-CoV entry. Reducing NUAK2 abundance or kinase activity impaired SARS-CoV-2 particle binding and internalization by decreasing cell surface levels of viral receptors and viral trafficking likely by modulating the actin cytoskeleton. IRE1α-dependent NUAK2 levels were elevated in SARS-CoV-2-infected and bystander non-infected cells, promoting viral spread by maintaining ACE2 cell surface levels and facilitating virion binding to bystander cells.


Asunto(s)
Proteínas Serina-Treonina Quinasas , SARS-CoV-2 , Internalización del Virus , Humanos , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/metabolismo , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , SARS-CoV-2/fisiología , Respuesta de Proteína Desplegada
2.
EMBO Rep ; 24(5): e57231, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37021481

RESUMEN

The cellular integrated stress response (ISR) is a central signaling pathway that tunes translation initiation in response to a wide range of cellular insults to promote cell survival. The critical node of this regulation involves the phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α) by stress kinases. In this issue of EMBO reports, Wu et al (2023) report FAM69C as a novel eIF2α kinase that promotes ISR activation and stress granule (SG) assembly in microglia in response to oxidative stress. This work proposes a protective role for FAM69C and SGs in limiting damaging inflammatory responses commonly associated with neurodegenerative diseases.


Asunto(s)
Microglía , eIF-2 Quinasa , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Microglía/metabolismo , Fosforilación , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Estrés Oxidativo
3.
J Virol ; 95(4)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33257477

RESUMEN

Positive-strand RNA viruses have been the etiological agents in several major disease outbreaks over the last few decades. Examples of this include flaviviruses, such as dengue virus and Zika virus, which cause millions of yearly infections around the globe, and coronaviruses, such as SARS-CoV-2, the source of the current pandemic. The severity of outbreaks caused by these viruses stresses the importance of research aimed at determining methods to limit virus spread and to curb disease severity. Such studies require molecular tools to decipher virus-host interactions and to develop effective treatments. Here, we describe the generation and characterization of a reporter system that can be used to visualize and identify cells infected with dengue virus or SARS-CoV-2. This system is based on viral protease activity that mediates cleavage and nuclear translocation of an engineered fluorescent protein stably expressed in cells. We show the suitability of this system for live cell imaging, for visualization of single infected cells, and for screening and testing of antiviral compounds. With the integrated modular building blocks, this system is easy to manipulate and can be adapted to any virus encoding a protease, thus offering a high degree of flexibility.IMPORTANCE Reporter systems are useful tools for fast and quantitative visualization of virus-infected cells within a host cell population. Here, we describe a reporter system that takes advantage of virus-encoded proteases expressed in infected cells to cleave an ER-anchored fluorescent protein fused to a nuclear localization sequence. Upon cleavage, the GFP moiety translocates to the nucleus, allowing for rapid detection of the infected cells. Using this system, we demonstrate reliable reporting activity for two major human pathogens from the Flaviviridae and the Coronaviridae families: dengue virus and SARS-CoV-2. We apply this reporter system to live cell imaging and use it for proof-of-concept to validate antiviral activity of a nucleoside analogue. This reporter system is not only an invaluable tool for the characterization of viral replication, but also for the discovery and development of antivirals that are urgently needed to halt the spread of these viruses.


Asunto(s)
COVID-19/virología , Virus del Dengue/aislamiento & purificación , Dengue/virología , SARS-CoV-2/aislamiento & purificación , Células A549 , Animales , COVID-19/diagnóstico , COVID-19/metabolismo , COVID-19/patología , Línea Celular , Chlorocebus aethiops , Dengue/diagnóstico , Dengue/metabolismo , Dengue/patología , Virus del Dengue/genética , Virus del Dengue/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Señales de Localización Nuclear/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Células Vero , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
4.
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
5.
EMBO Rep ; 21(12): e49019, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33180995

RESUMEN

Several human pathologies including neurological, cardiac, infectious, cancerous, and metabolic diseases have been associated with altered mitochondria morphodynamics. Here, we identify a small organic molecule, which we named Mito-C. Mito-C is targeted to mitochondria and rapidly provokes mitochondrial network fragmentation. Biochemical analyses reveal that Mito-C is a member of a new class of heterocyclic compounds that target the NEET protein family, previously reported to regulate mitochondrial iron and ROS homeostasis. One of the NEET proteins, NAF-1, is identified as an important regulator of mitochondria morphodynamics that facilitates recruitment of DRP1 to the ER-mitochondria interface. Consistent with the observation that certain viruses modulate mitochondrial morphogenesis as a necessary part of their replication cycle, Mito-C counteracts dengue virus-induced mitochondrial network hyperfusion and represses viral replication. The newly identified chemical class including Mito-C is of therapeutic relevance for pathologies where altered mitochondria dynamics is part of disease etiology and NEET proteins are highlighted as important therapeutic targets in anti-viral research.


Asunto(s)
Mitocondrias , Proteínas Mitocondriales , Homeostasis , Humanos , Hierro , Proteínas Mitocondriales/genética
6.
RNA Biol ; 18(5): 696-708, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33356825

RESUMEN

Beyond their high clinical relevance worldwide, flaviviruses (comprising dengue and Zika viruses) are of particular interest to understand the spatiotemporal control of RNA metabolism. Indeed, their positive single-stranded viral RNA genome (vRNA) undergoes in the cytoplasm replication, translation and encapsidation, three steps of the flavivirus life cycle that are coordinated through a fine-tuned equilibrium. Over the last years, RNA methylation has emerged as a powerful mechanism to regulate messenger RNA metabolism at the posttranscriptional level. Not surprisingly, flaviviruses exploit RNA epigenetic strategies to control crucial steps of their replication cycle as well as to evade sensing by the innate immune system. This review summarizes the current knowledge about vRNA methylation events and their impacts on flavivirus replication and pathogenesis. We also address the important challenges that the field of epitranscriptomics faces in reliably and accurately identifying RNA methylation sites, which should be considered in future studies on viral RNA modifications.


Asunto(s)
Epigénesis Genética/fisiología , Flavivirus/genética , ARN Viral/genética , Animales , Flavivirus/metabolismo , Flavivirus/patogenicidad , Genoma Viral , Humanos , Metilación , ARN Viral/metabolismo , Replicación Viral/genética , Virus Zika/genética , Virus Zika/metabolismo , Infección por el Virus Zika/genética , Infección por el Virus Zika/virología
7.
Bioorg Med Chem ; 47: 116392, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34509861

RESUMEN

In recent years, dengue virus (DENV) and Zika virus (ZIKV), both mosquito-borne members of the Flaviviridae family, have emerged as intercontinental health issues since their vectors have spread from their tropical origins to temperate climate zones due to climate change and increasing globalization. DENV and ZIKV are positive-sense, single-stranded RNA viruses, whose genomes consist of three structural (capsid, membrane precursor, envelope) and seven non-structural (NS) proteins, all of which are initially expressed as a single precursor polyprotein. For virus maturation, the polyprotein processing is accomplished by host proteases and the viral NS2B/NS3 protease complex, whose inhibitors have been shown to be effective antiviral agents with loss of viral pathogenicity. In this work, we elucidate new structure-activity relationships of benzo[d]thiazole-based allosteric NS2B/NS3 inhibitors. We developed a new series of Y-shaped inhibitors, which, with its larger hydrophobic contact surface, should bind to previously unaddressed regions of the allosteric NS2B/NS3 binding pocket. By scaffold-hopping, we varied the benzo[d]thiazole core and identified benzofuran as a new lead scaffold shifting the selectivity of initially ZIKV-targeting inhibitors to higher activities towards the DENV protease. In addition, we were able to increase the ligand efficiency from 0.27 to 0.41 by subsequent inhibitor truncation and identified N-(5,6-dihydroxybenzo[d]thiazol-2-yl)-4-iodobenzamide as a novel sub-micromolar NS2B/NS3 inhibitor. Utilizing cell-based assays, we could prove the antiviral activity in cellulo. Overall, we report new series of sub-micromolar allosteric DENV and ZIKV inhibitors with good efficacy profile in terms of cytotoxicity and protease inhibition selectivity.


Asunto(s)
Benzotiazoles/farmacología , Inhibidores de Proteasas/farmacología , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas Virales/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Benzotiazoles/síntesis química , Benzotiazoles/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Relación Estructura-Actividad , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/metabolismo
8.
J Immunol ; 200(11): 3857-3866, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29661824

RESUMEN

Dengue fever can be caused by one of four distinct dengue virus (DENV) serotypes that cocirculate in many parts of the world. Point of care serotype-specific nonstructural protein-1 (NS1) capture assays for the rapid serotyping of DENV in human sera would greatly support epidemiological surveillance and potentially also prognosis in individual patients. To ensure both serotype specificity and broad coverage of variants within serotypes, we have applied an innovative approach for the generation and selection of serotype-specific anti-NS1 mAbs. To elicit mAbs against conformational epitopes, NMRI mice were immunized with living HEK 293 transfectants expressing the native target Ags in multiple display on the cell surface. For each serotype, three different NS1 sequence variants were sequentially used for immunization of mice, hybridoma selection, and capture assay development, respectively. Selection of optimal combinations of capturing and detecting mAbs yielded highly sensitive and specific NS1 serotyping ELISAs (st-ELISAs) for the four serotypes. st-ELISA testing of 41 dengue patient sera showed a 100% concordance with the serotype determined by serotype-specific reverse transcriptase real-time quantitative PCR. The respective NS1 variants could be detected for ∼10 d after the onset of illness. Ab-dependent enhancement of DENV infections may be associated with a specific range of pre-existing anti-DENV serological Ab titers. Testing of patient sera with the developed st-ELISAs will not only be useful for epidemiological studies and surveillance, but it may also help to develop and validate assays that can distinguish protective versus enhancing Ab responses for risk assessment for the development of severe dengue disease in individual patients.


Asunto(s)
Virus del Dengue/inmunología , Serotipificación/métodos , Suero/inmunología , Suero/virología , Proteínas no Estructurales Virales/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Línea Celular , Reacciones Cruzadas/inmunología , Dengue/sangre , Dengue/inmunología , Dengue/virología , Epítopos/inmunología , Células HEK293 , Humanos , Inmunización/métodos , Sensibilidad y Especificidad , Serogrupo
9.
J Biol Chem ; 292(33): 13702-13713, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28659343

RESUMEN

The host-cell restriction factor SERINC5 potently suppresses the infectivity of HIV, type 1 (HIV-1) particles, and is counteracted by the viral pathogenesis factor Nef. However, the molecular mechanism by which SERINC5 restricts HIV-1 particle infectivity is still unclear. Because SERINC proteins have been suggested to facilitate the incorporation of serine during the biosynthesis of membrane lipids and because lipid composition of HIV particles is a major determinant of the infectious potential of the particles, we tested whether SERINC5-mediated restriction of HIV particle infectivity involves alterations of membrane lipid composition. We produced and purified HIV-1 particles from SERINC5293T cells with very low endogenous SERINC5 levels under conditions in which ectopically expressed SERINC5 restricts HIV-1 infectivity and is antagonized by Nef and analyzed both virions and producer cells with quantitative lipid MS. SERINC5 restriction and Nef antagonism were not associated with significant alterations in steady-state lipid composition of producer cells and HIV particles. Sphingosine metabolism kinetics were also unaltered by SERINC5 expression. Moreover, the levels of phosphatidylserine on the surface of HIV-1 particles, which may trigger uptake into non-productive internalization pathways in target cells, did not change upon expression of SERINC5 or Nef. Finally, saturating the phosphatidylserine-binding sites on HIV target cells did not affect SERINC5 restriction or Nef antagonism. These results demonstrate that the restriction of HIV-1 particle infectivity by SERINC5 does not depend on alterations in lipid composition and organization of HIV-1 particles and suggest that channeling serine into lipid biosynthesis may not be a cardinal cellular function of SERINC5.


Asunto(s)
VIH-1/patogenicidad , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Virión/patogenicidad , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Unión Competitiva , Línea Celular Tumoral , Eliminación de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , VIH-1/química , VIH-1/fisiología , Humanos , Cinética , Liposomas , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Esfingosina/metabolismo , Propiedades de Superficie , Virión/química , Virión/fisiología , Ensamble de Virus , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
10.
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
11.
J Virol ; 90(4): 2064-76, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26656698

RESUMEN

UNLABELLED: Production of proinflammatory cytokines indicative of potent recognition by the host innate immune system has long been recognized as a hallmark of the acute phase of HIV-1 infection. The first components of the machinery by which primary HIV target cells sense infection have recently been described; however, the mechanistic dissection of innate immune recognition and viral evasion would be facilitated by an easily accessible cell line model. Here we describe that reconstituted expression of the innate signaling adaptor STING enhanced the ability of the well-established HIV reporter cell line Tzm-bl to sense HIV infection and to convert this information into nuclear translocation of IRF3 as well as expression of cytokine mRNA. STING-dependent immune sensing of HIV-1 required virus entry and reverse transcription but not genome integration. Particularly efficient recognition was observed for an HIV-1 variant lacking expression of the accessory protein Vpr, suggesting a role of the viral protein in circumventing STING-mediated immune signaling. Vpr as well as STING significantly impacted the magnitude and breadth of the cytokine mRNA expression profile induced upon HIV-1 infection. However, cytoplasmic DNA sensing did not result in detectable cytokine secretion in this cell system, and innate immune recognition did not affect infection rates. Despite these deficits in eliciting antiviral effector functions, these results establish Tzm-bl STING and Tzm-bl STING IRF3.GFP cells as useful tools for studies aimed at dissecting mechanisms and regulation of early innate immune recognition of HIV infection. IMPORTANCE: Cell-autonomous immune recognition of HIV infection was recently established as an important aspect by which the host immune system attempts to fend off HIV-1 infection. Mechanistic studies on host cell recognition and viral evasion are hampered by the resistance of many primary HIV target cells to detailed experimental manipulation. We describe here that expression of the signaling adaptor STING renders the well-established HIV reporter cell line Tzm-bl competent for innate recognition of HIV infection. Key characteristics reflected in this cell model include nuclear translocation of IRF3, expression of a broad range of cytokine mRNAs, and an antagonistic activity of the HIV-1 protein Vpr. These results establish Tzm-bl STING and Tzm-bl STING IRF3.GFP cells as a useful tool for studies of innate recognition of HIV infection.


Asunto(s)
VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/biosíntesis , Línea Celular , Citocinas/biosíntesis , Citocinas/genética , Expresión Génica , Humanos , Evasión Inmune , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/genética , Transporte de Proteínas , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo
12.
PLoS Pathog ; 11(12): e1005345, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26720415

RESUMEN

Dengue virus (DENV) is the most common mosquito-transmitted virus infecting ~390 million people worldwide. In spite of this high medical relevance, neither a vaccine nor antiviral therapy is currently available. DENV elicits a strong interferon (IFN) response in infected cells, but at the same time actively counteracts IFN production and signaling. Although the kinetics of activation of this innate antiviral defense and the timing of viral counteraction critically determine the magnitude of infection and thus disease, quantitative and kinetic analyses are lacking and it remains poorly understood how DENV spreads in IFN-competent cell systems. To dissect the dynamics of replication versus antiviral defense at the single cell level, we generated a fully viable reporter DENV and host cells with authentic reporters for IFN-stimulated antiviral genes. We find that IFN controls DENV infection in a kinetically determined manner that at the single cell level is highly heterogeneous and stochastic. Even at high-dose, IFN does not fully protect all cells in the culture and, therefore, viral spread occurs even in the face of antiviral protection of naïve cells by IFN. By contrast, a vaccine candidate DENV mutant, which lacks 2'-O-methylation of viral RNA is profoundly attenuated in IFN-competent cells. Through mathematical modeling of time-resolved data and validation experiments we show that the primary determinant for attenuation is the accelerated kinetics of IFN production. This rapid induction triggered by mutant DENV precedes establishment of IFN-resistance in infected cells, thus causing a massive reduction of virus production rate. In contrast, accelerated protection of naïve cells by paracrine IFN action has negligible impact. In conclusion, these results show that attenuation of the 2'-O-methylation DENV mutant is primarily determined by kinetics of autocrine IFN action on infected cells.


Asunto(s)
Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Interferones/inmunología , Modelos Teóricos , Línea Celular , Supervivencia Celular , Vacunas contra el Dengue/genética , Virus del Dengue/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Metilación , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
PLoS Pathog ; 11(1): e1004573, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25569684

RESUMEN

Hepatitis C virus (HCV) is a major cause of chronic liver disease affecting around 130 million people worldwide. While great progress has been made to define the principle steps of the viral life cycle, detailed knowledge how HCV interacts with its host cells is still limited. To overcome this limitation we conducted a comprehensive whole-virus RNA interference-based screen and identified 40 host dependency and 16 host restriction factors involved in HCV entry/replication or assembly/release. Of these factors, heterogeneous nuclear ribonucleoprotein K (HNRNPK) was found to suppress HCV particle production without affecting viral RNA replication. This suppression of virus production was specific to HCV, independent from assembly competence and genotype, and not found with the related Dengue virus. By using a knock-down rescue approach we identified the domains within HNRNPK required for suppression of HCV particle production. Importantly, HNRNPK was found to interact specifically with HCV RNA and this interaction was impaired by mutations that also reduced the ability to suppress HCV particle production. Finally, we found that in HCV-infected cells, subcellular distribution of HNRNPK was altered; the protein was recruited to sites in close proximity of lipid droplets and colocalized with core protein as well as HCV plus-strand RNA, which was not the case with HNRNPK variants unable to suppress HCV virion formation. These results suggest that HNRNPK might determine efficiency of HCV particle production by limiting the availability of viral RNA for incorporation into virions. This study adds a new function to HNRNPK that acts as central hub in the replication cycle of multiple other viruses.


Asunto(s)
Hepacivirus/fisiología , Ribonucleoproteínas/fisiología , Virión/fisiología , Ensamble de Virus/genética , Células Cultivadas , Células HEK293 , Hepacivirus/efectos de los fármacos , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Humanos , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/farmacología , ARN Viral/metabolismo , Ribonucleoproteínas/antagonistas & inhibidores , Virión/efectos de los fármacos , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
14.
BMC Biotechnol ; 16(1): 83, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27876044

RESUMEN

BACKGROUND: Due to their rising incidence and progressive geographical spread, infections with mosquito-borne viruses, such as dengue (DENV), chikungunya and zika virus, have developed into major public health challenges. Since all of these viruses may cause similar symptoms and can occur in concurrent epidemics, tools for their differential diagnosis and epidemiological monitoring are of urgent need. RESULTS: Here we report the application of a novel strategy to rapidly generate monoclonal antibodies (mAbs) against native viral antigens, exemplified for the DENV nonstructural glycoprotein 1 (NS1). The described system is based on the immunization of mice with transfected mammalian cells expressing the target antigens in multiple displays on their cell surface and thereby presenting them efficiently to the host immune system in their native conformation. By applying this cell-based approach to the DENV NS1 protein of serotypes 1 (D1NS1) and 4 (D4NS1), we were able to rapidly generate panels of DENV NS1 serotype cross-reactive, as well as D1NS1- and D4NS1 serotype-specific mAbs. Our data show that the generated mAbs were capable of recognizing the endogenous NS1 protein in DENV-containing biological samples. CONCLUSION: The use of this novel immunization strategy, allows for a fast and efficient generation of hybridoma cell lines, producing mAbs against native viral antigens. Envisaged applications of the mAbs include the development of test platforms enabling a differentiation of the DENV serotypes and high resolution immunotyping for epidemiological studies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Presentadoras de Antígenos/inmunología , Antígenos Virales/inmunología , Evaluación Preclínica de Medicamentos/métodos , Inmunización/métodos , Proteínas no Estructurales Virales/inmunología , Animales , Diseño de Fármacos , Mapeo Epitopo , Células HEK293 , Humanos , Inmunoensayo/métodos , Ratones
15.
J Virol ; 89(20): 10548-68, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269178

RESUMEN

UNLABELLED: All major types of interferon (IFN) efficiently inhibit hepatitis C virus (HCV) replication in vitro and in vivo. Remarkably, HCV replication is not sensitive to IFN-γ in the hepatoma cell line Huh6, despite an intact signaling pathway. We performed transcriptome analyses between Huh6 and Huh-7 cells to identify effector genes of the IFN-γ response and thereby identified the DExD/H box helicase DEAD box polypeptide 60-like (DDX60L) as a restriction factor of HCV replication. DDX60L and its homolog DEAD box polypeptide 60 (DDX60) were both induced upon viral infection and IFN treatment in primary human hepatocytes. However, exclusively DDX60L knockdown increased HCV replication in Huh-7 cells and rescued HCV replication from type II IFN as well as type I and III IFN treatment, suggesting that DDX60L is an important effector protein of the innate immune response against HCV. In contrast, we found no impact of DDX60L on replication of hepatitis A virus. DDX60L protein was detectable only upon strong ectopic overexpression, displayed a broad cytoplasmic distribution, but caused cytopathic effects under these conditions. DDX60L knockdown did not alter interferon-stimulated gene (ISG) induction after IFN treatment but inhibited HCV replication upon ectopic expression, suggesting that it is a direct effector of the innate immune response. It most likely inhibits viral RNA replication, since we found neither impact of DDX60L on translation or stability of HCV subgenomic replicons nor additional impact on assembly of infectious virus. Similar to DDX60, DDX60L had a moderate impact on RIG-I dependent activation of innate immunity, suggesting additional functions in the sensing of viral RNA. IMPORTANCE: Interferons induce a plethora of interferon-stimulated genes (ISGs), which are our first line of defense against viral infections. In addition, IFNs have been used in antiviral therapy, in particular against the human pathogen hepatitis C virus (HCV); still, their mechanism of action is not well understood, since diverse, overlapping sets of antagonistic effector ISGs target viruses with different biologies. Our work identifies DDX60L as a novel factor that inhibits replication of HCV. DDX60L expression is regulated similarly to that of its homolog DDX60, but our data suggest that it has distinct functions, since we found no contribution of DDX60 in combatting HCV replication. The identification of novel components of the innate immune response contributes to a comprehensive understanding of the complex mechanisms governing antiviral defense.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Hepacivirus/genética , Hepatocitos/efectos de los fármacos , Interferón gamma/farmacología , Replicación Viral/genética , Línea Celular Tumoral , Supervivencia Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , Regulación de la Expresión Génica , Genes Reporteros , Genotipo , Hepacivirus/efectos de los fármacos , Hepacivirus/inmunología , Virus de la Hepatitis A/efectos de los fármacos , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/inmunología , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Luciferasas/genética , Luciferasas/inmunología , Cultivo Primario de Células , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Receptores Inmunológicos , Replicón , Transducción de Señal , Transcriptoma
16.
Nat Chem Biol ; 15(1): 5-6, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30531906
18.
J Hepatol ; 63(4): 829-37, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25908268

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) is a multifunctional protein playing a crucial role in diverse steps of the viral replication cycle and perturbing multiple host cell pathways. We showed previously that removal of a region in domain 2 (D2) of NS5A (mutant NS5A(D2Δ)) is dispensable for viral replication in hepatoma cell lines. By using a mouse model and immune-competent cell systems, we studied the role of D2 in controlling the innate immune response. METHODS: In vivo replication competence of NS5A(D2Δ) was studied in transgenic mice with human liver xenografts. Results were validated using primary human hepatocytes (PHHs) and mechanistic analyses were conducted in engineered Huh7 hepatoma cells with reconstituted innate signaling pathways. RESULTS: Although the deletion in NS5A removed most of the interferon (IFN) sensitivity determining-region, mutant NS5A(D2Δ) was as sensitive as the wild type to IFN-α and IFN-λ in vitro, but severely attenuated in vivo. This attenuation could be recapitulated in PHHs and was linked to higher activation of the IFN response, concomitant with reduced viral replication and virus production. Importantly, immune-reconstituted Huh7-derived cell lines revealed a sequential activation of the IFN-response via RIG-I (retinoic acid-inducible gene I) and MDA5 (Myeloma differentiation associated factor 5), respectively, that was significantly higher in the case of the mutant lacking most of NS5A D2. CONCLUSIONS: Our study reveals an important role of NS5A D2 for suppression of the IFN response that is activated by HCV via RIG-I and MDA5 in a sequential manner.


Asunto(s)
ADN Viral/genética , Hepacivirus/genética , Hepatitis C/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Mutación/genética , Proteínas no Estructurales Virales/genética , Animales , Antivirales/uso terapéutico , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Femenino , Genotipo , Hepacivirus/efectos de los fármacos , Hepatitis C/patología , Hepatitis C/virología , Hepatocitos , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas no Estructurales Virales/metabolismo
19.
PLoS Pathog ; 9(5): e1003359, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23675303

RESUMEN

The lipid kinase phosphatidylinositol 4-kinase III alpha (PI4KIIIα) is an essential host factor of hepatitis C virus (HCV) replication. PI4KIIIα catalyzes the synthesis of phosphatidylinositol 4-phosphate (PI4P) accumulating in HCV replicating cells due to enzyme activation resulting from its interaction with nonstructural protein 5A (NS5A). This study describes the interaction between PI4KIIIα and NS5A and its mechanistic role in viral RNA replication. We mapped the NS5A sequence involved in PI4KIIIα interaction to the carboxyterminal end of domain 1 and identified a highly conserved PI4KIIIα functional interaction site (PFIS) encompassing seven amino acids, which are essential for viral RNA replication. Mutations within this region were also impaired in NS5A-PI4KIIIα binding, reduced PI4P levels and altered the morphology of viral replication sites, reminiscent to the phenotype observed by silencing of PI4KIIIα. Interestingly, abrogation of RNA replication caused by mutations in the PFIS correlated with increased levels of hyperphosphorylated NS5A (p58), indicating that PI4KIIIα affects the phosphorylation status of NS5A. RNAi-mediated knockdown of PI4KIIIα or pharmacological ablation of kinase activity led to a relative increase of p58. In contrast, overexpression of enzymatically active PI4KIIIα increased relative abundance of basally phosphorylated NS5A (p56). PI4KIIIα therefore regulates the phosphorylation status of NS5A and viral RNA replication by favoring p56 or repressing p58 synthesis. Replication deficiencies of PFIS mutants in NS5A could not be rescued by increasing PI4P levels, but by supplying functional NS5A, supporting an essential role of PI4KIIIα in HCV replication regulating NS5A phosphorylation, thereby modulating the morphology of viral replication sites. In conclusion, we demonstrate that PI4KIIIα activity affects the NS5A phosphorylation status. Our results highlight the importance of PI4KIIIα in the morphogenesis of viral replication sites and its regulation by facilitating p56 synthesis.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Hepacivirus/fisiología , Interacciones Huésped-Patógeno/fisiología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Fosforilación , ARN Viral
20.
PLoS Pathog ; 9(7): e1003440, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23853584

RESUMEN

Influenza A NS1 and NS2 proteins are encoded by the RNA segment 8 of the viral genome. NS1 is a multifunctional protein and a virulence factor while NS2 is involved in nuclear export of viral ribonucleoprotein complexes. A yeast two-hybrid screening strategy was used to identify host factors supporting NS1 and NS2 functions. More than 560 interactions between 79 cellular proteins and NS1 and NS2 proteins from 9 different influenza virus strains have been identified. These interacting proteins are potentially involved in each step of the infectious process and their contribution to viral replication was tested by RNA interference. Validation of the relevance of these host cell proteins for the viral replication cycle revealed that 7 of the 79 NS1 and/or NS2-interacting proteins positively or negatively controlled virus replication. One of the main factors targeted by NS1 of all virus strains was double-stranded RNA binding domain protein family. In particular, adenosine deaminase acting on RNA 1 (ADAR1) appeared as a pro-viral host factor whose expression is necessary for optimal viral protein synthesis and replication. Surprisingly, ADAR1 also appeared as a pro-viral host factor for dengue virus replication and directly interacted with the viral NS3 protein. ADAR1 editing activity was enhanced by both viruses through dengue virus NS3 and influenza virus NS1 proteins, suggesting a similar virus-host co-evolution.


Asunto(s)
Adenosina Desaminasa/metabolismo , Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Replicación Viral , Adenosina Desaminasa/química , Adenosina Desaminasa/genética , Transporte Biológico , Línea Celular , Virus del Dengue/enzimología , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/patología , Gripe Humana/virología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Especificidad de la Especie , Técnicas del Sistema de Dos Híbridos , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA