11 research outputs found

    Cellular processing bodies and the hepatitis C virus life cycle : characterization of their dynamic interplay

    Get PDF
    Els cossos de processament (cossos-P) són grànuls discrets i dinàmics que contenen mRNAs represos en la traducció així com proteïnes involucrades en la degradació dels mRNAs i de la maquinària dels miRNAs. Alguns components dels cossos-P, com els repressors de la traducció PatL1, LSm1 i DDX6, promouen la traducció i la replicació del virus de la hepatitis C (VHC) i d’altres virus RNA de polaritat positiva [RNA(+)]. A més a més, un genome wide screening en llevat va determinar que l’exonucleasa Xrn1, que també es localitza als cossos-P, pot afectar la taxa de recombinació d’un virus RNA(+) de plantes Així doncs, els components dels cossos-P estan estretament relacionats amb els cicles vitals dels virus RNA(+). En l’estudi que aquí es presenta hem explorat la relació del VHC amb els cossos-P, demostrant que la infecció pel VHC promou canvis en la composició dels cossos-P a través de l’alteració de la localització d’aquells components que són necessaris per a la replicació viral. A més a més, hem demostrat que els components dels cossos-P però no els grànuls per se són necessaris per a la replicació del VHC. Addicionalment, hem posat a punt un sistema de detecció de recombinació en cultiu cel•lular basat en replicons del VHC que permet analitzar successos de recombinació i caracteritzar la possible participació dels components dels cossos-P en aquest mecanisme d’evolució. Amb aquest sistema s’han establert les primeres estimacions en la freqüència de recombinació del VHC indicant que la recombinació en aquest virus no és gaire comú. A més a més, la reducció del nivell d’expressió de Xrn1 no va alterar la taxa de recombinació del VHC indicant que la utilització de l’exonucleasa no és una característica general en la recombinació dels virus RNA(+). En conjunt, aquests resultats incrementen el nostre coneixement sobre els aspectes bàsics de la biologia del VHC així com de l’estreta relació que aquest virus estableix amb l’hoste

    Hepatitis C virus infection inhibits P-body granule formation in human livers

    No full text
    BACKGROUND & AIMS: Decoding the myriad of interactions that hepatitis C virus (HCV) establishes with infected cells is mandatory to obtain a complete understanding of HCV biology and its associated pathogenesis. We and others have previously found that HCV infection disrupts the formation of P-bodies in cell culture. These are cytoplasmic RNA granules with key roles in post-transcriptional regulation of gene expression. Therefore, P-body disruption might have consequences beyond viral propagation. However, whether P-body disruption occurs also in vivo is unknown. Aim of this study was to address this important issue. METHODS: Formalin-fixed paraffin-embedded liver biopsies from four groups of patients (healthy donors, patients with non-virus related liver inflammation, HCV- and HBV-infected patients) were immunostained to detect DDX6 and Dcp1, two core P-body components. Changes in the localization of these proteins were assessed by confocal microscopy. RESULTS: HCV specifically inhibited P-body formation in hepatocytes from human livers regardless of viral genotype, inflammation grade or whether the infection was recent or long established. Importantly, this alteration was reversed once HCV was eliminated by therapy. Furthermore, we observed in vivo an unexpected heterogeneity in P-body composition, which might reflect functional specializations. CONCLUSIONS: This is the first comprehensive in vivo P-body analysis that links a pathogenic condition to P-body alterations. Because of their role in gene expression, the alteration of P-bodies should be further studied to understand fully complex HCV-associated pathologies.JD and GPV were supported by a grant from the Spanish Ministry of Economy and Competitiveness (BFU2013-44629-R). XF received support in part by a grant from Instituto de Salud Carlos III (PI11/01907), Ministerio de Economía y Competitividad, cofunded by Fondo Europeo de Desarrollo Regional, Unión Europea, Una manera de hacer Europa. XF and SPP also received a grant/nfrom the Roche Organ Transplantation Research Foundation (ROTRF, CI: 442035057). The other authors were supported by grants from the following institutions: CFC from Asociación Española para el Estudio del Hígado, and LM from Instituto de Salud Carlos III, Ministerio de Economía y Competitivida

    Hepatitis C virus infection inhibits P-body granule formation in human livers

    No full text
    BACKGROUND & AIMS: Decoding the myriad of interactions that hepatitis C virus (HCV) establishes with infected cells is mandatory to obtain a complete understanding of HCV biology and its associated pathogenesis. We and others have previously found that HCV infection disrupts the formation of P-bodies in cell culture. These are cytoplasmic RNA granules with key roles in post-transcriptional regulation of gene expression. Therefore, P-body disruption might have consequences beyond viral propagation. However, whether P-body disruption occurs also in vivo is unknown. Aim of this study was to address this important issue. METHODS: Formalin-fixed paraffin-embedded liver biopsies from four groups of patients (healthy donors, patients with non-virus related liver inflammation, HCV- and HBV-infected patients) were immunostained to detect DDX6 and Dcp1, two core P-body components. Changes in the localization of these proteins were assessed by confocal microscopy. RESULTS: HCV specifically inhibited P-body formation in hepatocytes from human livers regardless of viral genotype, inflammation grade or whether the infection was recent or long established. Importantly, this alteration was reversed once HCV was eliminated by therapy. Furthermore, we observed in vivo an unexpected heterogeneity in P-body composition, which might reflect functional specializations. CONCLUSIONS: This is the first comprehensive in vivo P-body analysis that links a pathogenic condition to P-body alterations. Because of their role in gene expression, the alteration of P-bodies should be further studied to understand fully complex HCV-associated pathologies.JD and GPV were supported by a grant from the Spanish Ministry of Economy and Competitiveness (BFU2013-44629-R). XF received support in part by a grant from Instituto de Salud Carlos III (PI11/01907), Ministerio de Economía y Competitividad, cofunded by Fondo Europeo de Desarrollo Regional, Unión Europea, Una manera de hacer Europa. XF and SPP also received a grant/nfrom the Roche Organ Transplantation Research Foundation (ROTRF, CI: 442035057). The other authors were supported by grants from the following institutions: CFC from Asociación Española para el Estudio del Hígado, and LM from Instituto de Salud Carlos III, Ministerio de Economía y Competitivida

    CHIKV infection reprograms codon optimality to favor viral RNA translation by altering the tRNA epitranscriptome

    No full text
    Ample evidence indicates that codon usage bias regulates gene expression. How viruses, such as the emerging mosquito-borne Chikungunya virus (CHIKV), express their genomes at high levels despite an enrichment in rare codons remains a puzzling question. Using ribosome footprinting, we analyze translational changes that occur upon CHIKV infection. We show that CHIKV infection induces codon-specific reprogramming of the host translation machinery to favor the translation of viral RNA genomes over host mRNAs with an otherwise optimal codon usage. This reprogramming was mostly apparent at the endoplasmic reticulum, where CHIKV RNAs show high ribosome occupancy. Mechanistically, it involves CHIKV-induced overexpression of KIAA1456, an enzyme that modifies the wobble U34 position in the anticodon of tRNAs, which is required for proper decoding of codons that are highly enriched in CHIKV RNAs. Our findings demonstrate an unprecedented interplay of viruses with the host tRNA epitranscriptome to adapt the host translation machinery to viral production.This work was supported by the Spanish Ministry of Science and Innovation (PID2019-106959RB-I00/AEI/10.13039/501100011033 and PCIN-2016-106 to JD and PGC2018-098152-A-100 to EMN) and by an institutional “María de Maeztu” Programme for Units of Excellence in R&D (CEX2018-000792-M) and by the 2017 SGR 909 grant from the Secretaria d’Universitats i Recerca del Departament d’Economia i Coneixement de la Generalitat de Catalunya. RB was a recipient of a Juan de la Cierva fellowship. Mass spectrometric analyses were performed in the CRG/UPF Proteomics Unit (Proteored, PRB3, grant PT17/0019 PE I + D + i 2013-2016, ISCIII and ERDF). We thank C. V. Nicchitta and S. Leidel for experimental advice and F. Gebauer and A. Meyerhans for fruitful discussions. We acknowledge the support of the MEIC to the EMBL partnership, Centro de Excelencia Severo Ochoa and CERCA Programme/Generalitat de Catalunya

    Structural determinants of TRPV4 inhibition and identification of new antagonists with antiviral activity

    Get PDF
    Background and purpose: The transient receptor potential vanilloid 4 (TRPV4) cation channel participates in multiple physiological processes and is also at the core of different diseases, making this channel an interesting pharmacological target with therapeutic potential. However, little is known about the structural elements governing its inhibition. Experimental approach: We have now combined in silico drug discovery and molecular dynamics simulation based on Xenopus tropicalis xTRPV4 structure with functional studies measuring cell Ca2+ influx mediated by human TRPV4 channel to characterize the binding site of known TRPV4 inhibitors and to identify novel small molecule channel modulators. Key results: We have found that the inhibitor HC067047 binds to a pocket conformed by residues from S2-S3 linker (xTRPV4-D542), S4 (xTRPV4-M583 and Y587 and S5 (xTRPV4-D609 and F613). This pocket was also used for structure-based virtual screening in the search of novel channel modulators. Forty potential hits were selected based on the lower docking scores (from ~250,000 compounds) and their effect upon TRPV4 functionally tested. Three were further analysed for stability using molecular dynamics simulation and functionally tested on TRPV4 channels carrying mutations in the binding pocket. Compound NSC151066, shown to require residue xTRPV4-M583 for its inhibitory effect, presented an IC50 of 145 nM and demonstrated to be an effective antiviral against Zika virus with a potency similar to HC067047. Conclusion and implications: Together, we propose structural insights into the inhibition of TRPV4 and how this information can be used for the design of novel channel modulators.We thank Dr. Andres Merits (University of Tartu, Estonia) for kindly providing the plasmid encoding the ZIKV with NanoLuc, Spanish Ministry of Economy and Competitiveness through grants RTI2018‐099718 (to M.A.V.) and BFU2016‐80039‐R (to J.D.), an institutional “Unidad de Excelencia María de Maeztu” CEX2018‐000792‐M and FEDER funds. F.G.‐N. thanks Fondecyt Regular projects 1170733, the US Army of USA, W911NF‐14‐1‐0520 and The Centro Interdisciplinario de Neurociencia de Valparaíso is a Millennium Institute supported by the Millennium Scientific Initiative of the Ministerio de Economía, Fomento y Turismo P029‐022‐F

    Pan-genotypic hepatitis C virus inhibition by natural products derived from the wild Egyptian artichoke

    No full text
    Hepatitis C virus (HCV) infection is the leading cause of chronic liver diseases. Water extracts of the leaves of the wild Egyptian artichoke (WEA) [Cynara cardunculus L. var. sylvestris (Lam.) Fiori] have been used for centuries in the Sinai Peninsula to treat hepatitis symptoms. Here we isolated and characterized six compounds from the water extracts of WEA and evaluated their HCV inhibition capacities in vitro. Importantly, two of these compounds, grosheimol and cynaropicrin, inhibited HCV with half-maximal effective concentrations (EC50s) in the low micromolar range. They inhibited HCV entry into target cells and were active against both cell-free infection as well as cell-cell transmission. Furthermore, the antiviral activity of both compounds was pan-genotypic as HCV genotypes 1a, 1b, 2b, 3a, 4a, 5a, 6a, and 7a were inhibited. Thus, grosheimol and cynaropicrin are promising candidates for the development of new pan-genotypic entry inhibitors of HCV infection. IMPORTANCE:/nBecause there is no preventive HCV vaccine available today, the discovery of novel anti-HCV cell entry inhibitors could help develop preventive measures against infection. The present study describes two compounds isolated from the wild Egyptian artichoke (WEA) with respect to their structural elucidation, absolute configuration, and quantitative determination. Importantly, both compounds inhibited HCV infection in vitro. The first compound was an unknown molecule, and it was designated "grosheimol," while the second compound is the known molecule cynaropicrin. Both compounds belong to the group of sesquiterpene lactones. The mode of action of these compounds occurred during the early steps of the HCV life cycle, including cell-free and cell-cell infection inhibition. These natural compounds present promising candidates for further development into anti-HCV therapeutics.This work was supported by FEDER and the Spanish Ministry of Economy and Competitiveness through grants BFU 2013-44629-R and SAF2013-46077-R and the Maria de Maeztu Programme for Units of Excellence in R&D (MDM-2014-0370

    The TRPV4 channel links calcium influx to DDX3X activity and viral infectivity

    No full text
    Ion channels are well placed to transduce environmental cues into signals used by cells to generate a wide range of responses, but little is known about their role in the regulation of RNA metabolism. Here we show that the TRPV4 cation channel binds the DEAD-box RNA helicase DDX3X and regulates its function. TRPV4-mediated Ca2+ influx releases DDX3X from the channel and drives DDX3X nuclear translocation, a process that involves calmodulin (CaM) and the CaM-dependent kinase II. Genetic depletion or pharmacological inhibition of TRPV4 diminishes DDX3X-dependent functions, including nuclear viral export and translation. Furthermore, TRPV4 mediates Ca2+ influx and nuclear accumulation of DDX3X in cells exposed to the Zika virus or the purified viral envelope protein. Consequently, targeting of TRPV4 reduces infectivity of dengue, hepatitis C and Zika viruses. Together, our results highlight the role of TRPV4 in the regulation of DDX3X-dependent control of RNA metabolism and viral infectivity.This work was supported by the Spanish Ministry of Economy and Competitiveness through grants SAF2015-69762R, BFU2016-80039-R, BFU2017-87843-R, an institutional “Maria de Maeztu” Programme for Units of Excellence in R&D (MDM-2014-0370) and FEDER funds; Marie Curie International Outgoing Fellowship within the 7th European Community Framework Programme (PIOF-GA-2009-237120) and the Generalitat de Catalunya research program (AGAUR, 2014-SGR-1628 and FI-2013FIB00251)

    Host-derived circular RNAs display proviral activities in Hepatitis C virus-infected cells

    No full text
    Viruses subvert macromolecular pathways in infected host cells to aid in viral gene amplification or to counteract innate immune responses. Roles for host-encoded, noncoding RNAs, including microRNAs, have been found to provide pro- and anti-viral functions. Recently, circular RNAs (circRNAs), that are generated by a nuclear back-splicing mechanism of pre-mRNAs, have been implicated to have roles in DNA virus-infected cells. This study examines the circular RNA landscape in uninfected and hepatitis C virus (HCV)-infected liver cells. Results showed that the abundances of distinct classes of circRNAs were up-regulated or down-regulated in infected cells. Identified circRNAs displayed pro-viral effects. One particular up-regulated circRNA, circPSD3, displayed a very pronounced effect on viral RNA abundances in both hepatitis C virus- and Dengue virus-infected cells. Though circPSD3 has been shown to bind factor eIF4A3 that modulates the cellular nonsense-mediated decay (NMD) pathway, circPSD3 regulates RNA amplification in a pro-viral manner at a post-translational step, while eIF4A3 exhibits the anti-viral property of the NMD pathway. Findings from the global analyses of the circular RNA landscape argue that pro-, and likely, anti-viral functions are executed by circRNAs that modulate viral gene expression as well as host pathways. Because of their long half-lives, circRNAs likely play hitherto unknown, important roles in viral pathogenesis.This study was funded by the National Institutes of Health (https://www.nih.gov) (R01AI06900011) and NIH grants R01GM122406 and R01AG057700 to S.K. Funds for J.D. were received from the Spanish Ministry of Economy and Competitiveness (AEI/ MINECO/FEDER,UE) through grants BFU2016–80039-R and Unidad de Excelencia Maria de Maeztu funded by the MINECO (ref: MDM-2014–0370), and by a 2017 SGR 909 grant by the Secretaria d’Universitats i Recerca del Departament d’Economia i Coneixement of the Generalitat de Cataluny

    Oligonucleotide-lipid conjugates forming G-quadruplex structures are potent and pangenotypic hepatitis C virus entry inhibitors in vitro and ex vivo

    No full text
    A hepatitis C virus (HCV) epidemic affecting HIV-infected men who have sex with men (MSM) is expanding worldwide. In spite of the improved cure rates obtained with the new direct-acting antiviral drug (DAA) combinations, the high rate of reinfection within this population calls urgently for novel preventive interventions. In this study, we determined in cell culture and ex vivo experiments with human colorectal tissue that lipoquads, G-quadruplex DNA structures fused to cholesterol, are efficient HCV pangenotypic entry and cell-to-cell transmission inhibitors. Thus, lipoquads may be promising candidates for the development of rectally applied gels to prevent HCV transmission.The authors are supported by grants BFU2013-44629-R (to JD, GK and APL), and SAF2013-46077-R (to AM), and also through the “María de Maeztu” Programme for Units of Excellence in R&D (MDM-2014-0370), all from the Spanish Ministry of Economy and Competitiveness and FEDER. Work at Imperial College (CH) was funded by EC FP7 grant 305578 (PathCO)

    Soraphen A: a broad-spectrum antiviral natural product with potent anti-hepatitis C virus activity

    No full text
    BACKGROUND & AIMS: Soraphen A (SorA) is a myxobacterial metabolite that inhibits the acetyl-CoA carboxylase, a key enzyme in lipid biosynthesis. We have previously identified SorA to efficiently inhibit the human immunodeficiency virus (HIV). The aim of the present study was to evaluate the capacity of SorA and analogues to inhibit hepatitis C virus (HCV) infection. METHODS: SorA inhibition capacity was evaluated in vitro using cell culture derived HCV, HCV pseudoparticles and subgenomic replicons. Infection studies were performed in the hepatoma cell line HuH7/Scr and in primary human hepatocytes. The effects of SorA on membranous web formation were analysed by electron microscopy./nRESULTS:/nSorA potently inhibits HCV infection at nanomolar concentrations. Obtained EC50 values were 0.70 nM with a HCV reporter genome, 2.30 nM with wild-type HCV and 2.52 nM with subgenomic HCV replicons. SorA neither inhibited HCV RNA translation nor HCV entry, as demonstrated with subgenomic HCV replicons and HCV pseudoparticles, suggesting an effect on HCV replication. Consistent with this, evidence was obtained that SorA interferes with formation of the membranous web, the site of HCV replication. Finally, a series of natural and synthetic SorA analogues helped to establish a first structure-activity relationship./nCONCLUSIONS:/nSorA has a very potent anti-HCV activity. Since it also interferes with the membranous web formation, SorA is an excellent tool to unravel the mechanism of HCV replication.The authors are supported by grants BFU2013-44629-R (to JD, GK and GPV), SAF2013-46077-R (to/nAM and JM), both from the Spanish Ministry of Economy and Competitiveness and FEDER, and from the German Centre for Infection Research (DZIF, to MB and RM
    corecore