Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 77
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
PLoS Pathog ; 16(9): e1008920, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32997730

RÉSUMÉ

The virions of enteroviruses such as poliovirus undergo a global conformational change after binding to the cellular receptor, characterized by a 4% expansion, and by the opening of holes at the two and quasi-three-fold symmetry axes of the capsid. The resultant particle is called a 135S particle or A-particle and is thought to be on the pathway to a productive infection. Previously published studies have concluded that the membrane-interactive peptides, namely VP4 and the N-terminus of VP1, are irreversibly externalized in the 135S particle. However, using established protocols to produce the 135S particle, and single particle cryo-electron microscopy methods, we have identified at least two unique states that we call the early and late 135S particle. Surprisingly, only in the "late" 135S particles have detectable levels of the VP1 N-terminus been trapped outside the capsid. Moreover, we observe a distinct density inside the capsid that can be accounted for by VP4 that remains associated with the genome. Taken together our results conclusively demonstrate that the 135S particle is not a unique conformation, but rather a family of conformations that could exist simultaneously.


Sujet(s)
Capside/ultrastructure , Poliomyélite/métabolisme , ARN viral/ultrastructure , Virion/ultrastructure , Capside/métabolisme , Protéines de capside/métabolisme , Cryomicroscopie électronique , Humains , Modèles moléculaires , ARN viral/métabolisme , Récepteurs viraux/métabolisme , Virion/métabolisme , Pénétration virale
2.
Virology ; 545: 53-62, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32308198

RÉSUMÉ

Viruses have evolved strategies to ensure efficient translation using host cell ribosomes and translation factors. In addition to cleaving translation initiation factors required for host cell translation, poliovirus (PV) uses an internal ribosome entry site (IRES). Recent studies suggest that viruses exploit specific ribosomal proteins to enhance translation of their viral proteins. The ribosomal protein receptor for activated C kinase 1 (RACK1), a protein of the 40S ribosomal subunit, was previously shown to mediate translation from the 5' cricket paralysis virus and hepatitis C virus IRESs. Here we found that translation of a PV dual-luciferase reporter shows a moderate dependence on RACK1. However, in the context of a viral infection we observed significantly reduced poliovirus plaque size and titers and delayed host cell translational shut-off. Our findings further illustrate the involvement of the cellular translational machinery during PV infection and how viruses usurp the function of specific ribosomal proteins.


Sujet(s)
Hepacivirus/génétique , Hépatite C/métabolisme , Sites internes d'entrée des ribosomes , Poliomyélite/métabolisme , Poliovirus/génétique , Récepteurs de kinase-C activée/métabolisme , Hepacivirus/métabolisme , Hépatite C/génétique , Hépatite C/virologie , Interactions hôte-pathogène , Humains , Poliomyélite/génétique , Poliomyélite/virologie , Poliovirus/métabolisme , Biosynthèse des protéines , Récepteurs de kinase-C activée/génétique , Ribosomes/génétique , Ribosomes/métabolisme
3.
J Virol ; 93(21)2019 11 01.
Article de Anglais | MEDLINE | ID: mdl-31375590

RÉSUMÉ

The replication of many positive-strand RNA viruses [(+)RNA viruses] depends on the cellular protein GBF1, but its role in the replication process is not clear. In uninfected cells, GBF1 activates small GTPases of the Arf family and coordinates multiple steps of membrane metabolism, including functioning of the cellular secretory pathway. The nonstructural protein 3A of poliovirus and related viruses has been shown to directly interact with GBF1, likely mediating its recruitment to the replication complexes. Surprisingly, viral mutants with a severely reduced level of 3A-GBF1 interaction demonstrate minimal replication defects in cell culture. Here, we systematically investigated the conserved elements of GBF1 to understand which determinants are important to support poliovirus replication. We demonstrate that multiple GBF1 mutants inactive in cellular metabolism could still be fully functional in the replication complexes. Our results show that the Arf-activating property, but not the primary structure of the Sec7 domain, is indispensable for viral replication. They also suggest a redundant mechanism of recruitment of GBF1 to the replication sites, which is dependent not only on direct interaction of the protein with the viral protein 3A but also on determinants located in the noncatalytic C-terminal domains of GBF1. Such a double-targeting mechanism explains the previous observations of the remarkable tolerance of different levels of GBF1-3A interaction by the virus and likely constitutes an important element of the resilience of viral replication.IMPORTANCE Enteroviruses are a vast group of viruses associated with diverse human diseases, but only two of them could be controlled with vaccines, and effective antiviral therapeutics are lacking. Here, we investigated in detail the contribution of a cellular protein, GBF1, in the replication of poliovirus, a representative enterovirus. GBF1 supports the functioning of cellular membrane metabolism and is recruited to viral replication complexes upon infection. Our results demonstrate that the virus requires a limited subset of the normal GBF1 functions and reveal the elements of GBF1 essential to support viral replication under different conditions. Since diverse viruses often rely on the same cellular proteins for replication, understanding the mechanisms by which these proteins support infection is essential for the development of broad-spectrum antiviral therapeutics.


Sujet(s)
Facteurs d'échange de nucléotides guanyliques/métabolisme , Poliovirus/physiologie , Réplication virale , Facteur-1 d'ADP-ribosylation/métabolisme , Protéines d'activation de la GTPase/génétique , Facteurs d'échange de nucléotides guanyliques/composition chimique , Facteurs d'échange de nucléotides guanyliques/génétique , Cellules HeLa , Interactions hôte-pathogène , Humains , Mutation , Poliomyélite/métabolisme , Poliomyélite/virologie , Poliovirus/métabolisme , Liaison aux protéines , Domaines protéiques , Protéines du core viral/métabolisme
4.
PLoS Pathog ; 14(4): e1007036, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29702686

RÉSUMÉ

At the culmination of poliovirus (PV) multiplication, membranes are observed that contain phosphatidylinositol-4-phosphate (PI4P) and appear as vesicular clusters in cross section. Induction and remodeling of PI4P and membranes prior to or concurrent with genome replication has not been well studied. Here, we exploit two PV mutants, termed EG and GG, which exhibit aberrant proteolytic processing of the P3 precursor that substantially delays the onset of genome replication and/or impairs virus assembly, to illuminate the pathway of formation of PV-induced membranous structures. For WT PV, changes to the PI4P pool were observed as early as 30 min post-infection. PI4P remodeling occurred even in the presence of guanidine hydrochloride, a replication inhibitor, and was accompanied by formation of membrane tubules throughout the cytoplasm. Vesicular clusters appeared in the perinuclear region of the cell at 3 h post-infection, a time too slow for these structures to be responsible for genome replication. Delays in the onset of genome replication observed for EG and GG PVs were similar to the delays in virus-induced remodeling of PI4P pools, consistent with PI4P serving as a marker of the genome-replication organelle. GG PV was unable to convert virus-induced tubules into vesicular clusters, perhaps explaining the nearly 5-log reduction in infectious virus produced by this mutant. Our results are consistent with PV inducing temporally distinct membranous structures (organelles) for genome replication (tubules) and virus assembly (vesicular clusters). We suggest that the pace of formation, spatiotemporal dynamics, and the efficiency of the replication-to-assembly-organelle conversion may be set by both the rate of P3 polyprotein processing and the capacity for P3 processing to yield 3AB and/or 3CD proteins.


Sujet(s)
Membrane cellulaire/composition chimique , Organites/virologie , Phosphates phosphatidylinositol/métabolisme , Poliomyélite/virologie , Poliovirus/pathogénicité , Protéines virales/métabolisme , Réplication virale , Membrane cellulaire/métabolisme , Génome viral , Cellules HeLa , Humains , Mutation , Phosphates phosphatidylinositol/composition chimique , Poliomyélite/génétique , Poliomyélite/métabolisme , Poliovirus/génétique , Analyse spatio-temporelle , Protéines virales/génétique , Assemblage viral
5.
J Appl Microbiol ; 124(4): 1001-1007, 2018 Apr.
Article de Anglais | MEDLINE | ID: mdl-29078036

RÉSUMÉ

AIMS: Cellular responses of an established cell line from human intestinal epithelial cells (INT-407 cells) against poliovirus (PV) infections were investigated in order to find cellular genetic markers for infectious PV detection. METHODS AND RESULTS: Gene expression profile of INT-407 cells was analysed by DNA microarray technique when cells were infected with poliovirus 1 (PV1) (sabin) at multiplicity of infection of 10-3 and incubated for 12 h. Poliovirus infection significantly altered the gene expressions of two ion channels, KCNJ4 and SCN7A. The expression profile of KCNJ4 gene was further investigated by real-time RT-qPCR, and it was found that KCNJ4 gene was significantly regulated at 24 h postinfection of PV1. CONCLUSIONS: KCNJ4 gene, coding a potassium channel protein, is proposed as a cellular genetic marker for infectious PV detection. SIGNIFICANCE AND IMPACT OF THE STUDY: This is the first study to show the availability of cellular responses to detect infectious PV. The selection of cellular genetic markers for infectious viruses using DNA microarray and RT-qPCR can be applicable for the other enteric viruses.


Sujet(s)
Poliomyélite/génétique , Poliovirus/isolement et purification , Lignée cellulaire , Expression des gènes , Marqueurs génétiques , Humains , Poliomyélite/métabolisme , Poliomyélite/virologie , Poliovirus/génétique , Poliovirus/physiologie , Canaux potassiques rectifiants entrants/génétique , Canaux potassiques rectifiants entrants/métabolisme , Canaux sodiques voltage-dépendants/génétique , Canaux sodiques voltage-dépendants/métabolisme
6.
Sci Rep ; 7(1): 10496, 2017 09 05.
Article de Anglais | MEDLINE | ID: mdl-28874814

RÉSUMÉ

Intracerebral Theiler's murine encephalomyelitis virus (TMEV) infection in mice induces inflammatory demyelination in the central nervous system. Although C57BL/6 mice normally resistant to TMEV infection with viral clearance, we have previously demonstrated that RORγt-transgenic (tg) C57BL/6 mice, which have Th17-biased responses due to RORγt overexpression in T cells, became susceptible to TMEV infection with viral persistence. Here, using T-bet-tg C57BL/6 mice and Gata3-tg C57BL/6 mice, we demonstrated that overexpression of T-bet, but not Gata3, in T cells was detrimental in TMEV infection. Unexpectedly, T-bet-tg mice died 2 to 3 weeks after infection due to failure of viral clearance. Here, TMEV infection induced splenic T cell depletion, which was associated with lower anti-viral antibody and T cell responses. In contrast, Gata3-tg mice remained resistant, while Gata3-tg mice had lower IFN-γ and higher IL-4 production with increased anti-viral IgG1 responses. Thus, our data identify how overexpression of T-bet and Gata3 in T cells alters anti-viral immunity and confers susceptibility to TMEV infection.


Sujet(s)
Facteur de transcription GATA-3/génétique , Expression des gènes , Poliomyélite/génétique , Poliomyélite/virologie , Protéines à domaine boîte-T/génétique , Theilovirus/physiologie , Animaux , Marqueurs biologiques , Cytokines/métabolisme , Maladies démyélinisantes/génétique , Prédisposition aux maladies , Facteur de transcription GATA-3/métabolisme , Interactions hôte-pathogène/immunologie , Immunohistochimie , Souris , Souris transgéniques , Poliomyélite/immunologie , Poliomyélite/métabolisme , Rate/immunologie , Rate/métabolisme , Protéines à domaine boîte-T/métabolisme , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Virulence , Réplication virale
7.
J Biol Chem ; 292(25): 10664-10671, 2017 06 23.
Article de Anglais | MEDLINE | ID: mdl-28446605

RÉSUMÉ

Several groups have used genome-wide libraries of lentiviruses encoding small guide RNAs (sgRNAs) for genetic screens. In most cases, sgRNA expression cassettes are integrated into cells by using lentiviruses, and target genes are statistically estimated by the readout of sgRNA sequences after targeted sequencing. We present a new virus-free method for human gene knockout screens using a genome-wide library of CRISPR/Cas9 sgRNAs based on plasmids and target gene identification via whole-genome sequencing (WGS) confirmation of authentic mutations rather than statistical estimation through targeted amplicon sequencing. We used 30,840 pairs of individually synthesized oligonucleotides to construct the genome-scale sgRNA library, collectively targeting 10,280 human genes (i.e. three sgRNAs per gene). These plasmid libraries were co-transfected with a Cas9-expression plasmid into human cells, which were then treated with cytotoxic drugs or viruses. Only cells lacking key factors essential for cytotoxic drug metabolism or viral infection were able to survive. Genomic DNA isolated from cells that survived these challenges was subjected to WGS to directly identify CRISPR/Cas9-mediated causal mutations essential for cell survival. With this approach, we were able to identify known and novel genes essential for viral infection in human cells. We propose that genome-wide sgRNA screens based on plasmids coupled with WGS are powerful tools for forward genetics studies and drug target discovery.


Sujet(s)
Clustered regularly interspaced short palindromic repeats , Génome humain , Séquençage nucléotidique à haut débit , Poliomyélite/génétique , Poliovirus , Techniques de knock-down de gènes , Étude d'association pangénomique , Cellules HeLa , Humains , Poliomyélite/métabolisme
8.
Nat Methods ; 14(1): 49-52, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27869813

RÉSUMÉ

We engineered covalently circularized nanodiscs (cNDs) which, compared with standard nanodiscs, exhibit enhanced stability, defined diameter sizes and tunable shapes. Reconstitution into cNDs enhanced the quality of nuclear magnetic resonance spectra for both VDAC-1, a ß-barrel membrane protein, and the G-protein-coupled receptor NTR1, an α-helical membrane protein. In addition, we used cNDs to visualize how simple, nonenveloped viruses translocate their genomes across membranes to initiate infection.


Sujet(s)
Double couche lipidique/composition chimique , Nanostructures/composition chimique , Récepteur neurotensine/métabolisme , Canal anionique-1 voltage-dépendant/métabolisme , Humains , Double couche lipidique/métabolisme , Modèles moléculaires , Résonance magnétique nucléaire biomoléculaire , Poliomyélite/métabolisme , Poliomyélite/virologie , Poliovirus/physiologie , Pénétration virale
9.
Glia ; 64(1): 105-21, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26337870

RÉSUMÉ

Multiple sclerosis (MS) frequently starts near the lateral ventricles, which are lined by subventricular zone (SVZ) progenitor cells that can migrate to lesions and contribute to repair. Because MS-induced inflammation may decrease SVZ proliferation and thus limit repair, we studied the role of galectin-3 (Gal-3), a proinflammatory protein. Gal-3 expression was increased in periventricular regions of human MS in post-mortem brain samples and was also upregulated in periventricular regions in a murine MS model, Theiler's murine encephalomyelitis virus (TMEV) infection. Whereas TMEV increased SVZ chemokine (CCL2, CCL5, CCL, and CXCL10) expression in wild type (WT) mice, this was inhibited in Gal-3(-/-) mice. Though numerous CD45+ immune cells entered the SVZ of WT mice after TMEV infection, their numbers were significantly diminished in Gal-3(-/-) mice. TMEV also reduced neuroblast and proliferative SVZ cell numbers in WT mice but this was restored in Gal-3(-/-) mice and was correlated with increased numbers of doublecortin+ neuroblasts in the corpus callosum. In summary, our data showed that loss of Gal-3 blocked chemokine increases after TMEV, reduced immune cell migration into the SVZ, reestablished SVZ proliferation and increased the number of progenitors in the corpus callosum. These results suggest Gal-3 plays a central role in modulating the SVZ neurogenic niche's response to this model of MS.


Sujet(s)
Encéphale/métabolisme , Galectine -3/métabolisme , Sclérose en plaques/métabolisme , Maladie neurologique auto-immune expérimentale/métabolisme , Neurogenèse , Niche de cellules souches/physiologie , Adolescent , Adulte , Sujet âgé , Animaux , Encéphale/immunologie , Encéphale/anatomopathologie , Mouvement cellulaire , Enfant , Femelle , Galectine -3/génétique , Humains , Mâle , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Sclérose en plaques/immunologie , Sclérose en plaques/anatomopathologie , Maladie neurologique auto-immune expérimentale/immunologie , Maladie neurologique auto-immune expérimentale/anatomopathologie , Cellules souches neurales/métabolisme , Cellules souches neurales/anatomopathologie , Poliomyélite/métabolisme , Poliomyélite/anatomopathologie , Theilovirus , Jeune adulte
10.
J Biol Chem ; 289(7): 3936-49, 2014 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-24382890

RÉSUMÉ

We have shown previously that poliovirus infection disrupts cytoplasmic P-bodies in infected mammalian cells. During the infectious cycle, poliovirus causes the directed cleavage of Dcp1a and Pan3, coincident with the dispersion of P-bodies. We now show that expression of Dcp1a prior to infection, surprisingly, restricts poliovirus infection. This inhibition of infection was independent of P-body formation because expression of GFP-Dcp1a mutants that cannot enter P-bodies restricted poliovirus infection similar to wild-type GFP-Dcp1a. Expression of wild-type or mutant GFP-Dcp1a induced phosphorylation of eIF2α through the eIF2α kinase protein kinase R (PKR). Activation of PKR required the amino-terminal EVH1 domain of Dcp1a. This PKR-induced translational inhibition appears to be specific to Dcp1a because the expression of other P-body components, Pan2, Pan3, Ccr4, or Caf1, did not result in the inhibition of poliovirus gene expression or induce eIF2α phosphorylation. The translation blockade induced by Dcp1a expression suggests novel signaling linking RNA degradation/decapping and regulation of translation.


Sujet(s)
Endoribonucleases/métabolisme , Biosynthèse des protéines/physiologie , Stabilité de l'ARN/physiologie , Transactivateurs/métabolisme , eIF-2 Kinase/métabolisme , Animaux , Lignée cellulaire , Endoribonucleases/génétique , Activation enzymatique/génétique , Facteur-2 d'initiation eucaryote/génétique , Facteur-2 d'initiation eucaryote/métabolisme , Exoribonucleases , Souris , Souris knockout , Mutation , Phosphorylation/génétique , Poliomyélite/génétique , Poliomyélite/métabolisme , Poliomyélite/anatomopathologie , Poliovirus/génétique , Poliovirus/métabolisme , Structure tertiaire des protéines , Protéines/génétique , Protéines/métabolisme , Récepteurs CCR4/génétique , Récepteurs CCR4/métabolisme , Protéines de répression , Ribonucléases , Transactivateurs/génétique , eIF-2 Kinase/génétique
11.
Cell Host Microbe ; 15(1): 36-46, 2014 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-24439896

RÉSUMÉ

Enteric viruses, including poliovirus and reovirus, encounter a vast microbial community in the mammalian gastrointestinal tract, which has been shown to promote virus replication and pathogenesis. Investigating the underlying mechanisms, we find that poliovirus binds bacterial surface polysaccharides, which enhances virion stability and cell attachment by increasing binding to the viral receptor. Additionally, we identified a poliovirus mutant, VP1-T99K, with reduced lipopolysaccharide (LPS) binding. Although T99K and WT poliovirus cell attachment, replication, and pathogenesis in mice are equivalent, VP1-T99K poliovirus was unstable in feces following peroral inoculation of mice. Consequently, the ratio of mutant virus in feces is reduced following additional cycles of infection in mice. Thus, the mutant virus incurs a fitness cost when environmental stability is a factor. These data suggest that poliovirus binds bacterial surface polysaccharides, enhancing cell attachment and environmental stability, potentially promoting transmission to a new host.


Sujet(s)
Interactions hôte-pathogène , Lipopolysaccharides/métabolisme , Interactions microbiennes/génétique , Poliomyélite/virologie , Poliovirus/métabolisme , Virion/métabolisme , Animaux , Lignée cellulaire tumorale , Fèces/virologie , Fibroblastes/virologie , Aptitude génétique/physiologie , Cellules HeLa , Humains , Souris , Souris transgéniques , Mutation , Poliomyélite/métabolisme , Poliomyélite/mortalité , Poliovirus/génétique , Poliovirus/pathogénicité , Liaison aux protéines , Récepteurs viraux/métabolisme , Analyse de survie , Méthode des plages virales , Virion/génétique , Virion/pathogénicité , Attachement viral , Réplication virale
12.
J Rehabil Med ; 46(1): 67-72, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24162727

RÉSUMÉ

OBJECTIVE: The aim of this study was to investigate signs of inflammation in muscle of patients with prior polio, since the main symptoms in these patients are muscle pain, weakness and fatigue. In the context of pain and inflammation, the prostaglandin E2 pathway is of interest. Prostaglandin E2 has many biological actions and is a mediator of inflammation and pain. PATIENTS AND METHODS: Skeletal muscle biopsies from 8 patients with prior polio and post-polio symptoms, presenting with pain and muscular weakness, and from 6 healthy controls were studied. Immunohistochemistry, conventional microscopy, and computerized image analysis were performed. RESULTS: There was statistically significant higher expression of enzymes of the prostaglandin E2 synthetic pathway, in muscle from patients, compared with controls. Expression of prostaglandin enzymes was mainly in scattered cells and blood vessels, and may indicate an inflammatory process of the muscle, which could be secondary to systemic inflammation. CONCLUSION: This data may indicate an inflammatory process in muscle of prior polio patients. Up-regulation of the prostaglandin E2 pathway reveals a potential background to the pain experienced by these patients, and may provide opportunities for directed pharmacological and physical therapies, which could lead to better outcomes of rehabilitation interventions.


Sujet(s)
Dinoprostone/métabolisme , Muscles squelettiques/métabolisme , Poliomyélite/métabolisme , Adulte , Dinoprostone/biosynthèse , Fatigue/métabolisme , Femelle , Humains , Mâle , Adulte d'âge moyen , Faiblesse musculaire/métabolisme , Myalgie/métabolisme , Syndrome post-poliomyélitique/métabolisme , Jeune adulte
13.
J Virol ; 88(6): 3192-201, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24371074

RÉSUMÉ

UNLABELLED: Due to their small genome size, picornaviruses must utilize host proteins to mediate cap-independent translation and viral RNA replication. The host RNA-binding protein poly(rC) binding protein 2 (PCBP2) is involved in both processes in poliovirus infected cells. It has been shown that the viral proteinase 3CD cleaves PCBP2 and contributes to viral translation inhibition. However, cleaved PCBP2 remains active in viral RNA replication. This would suggest that both cleaved and intact forms of PCBP2 have a role in the viral RNA replication cycle. The picornavirus genome must act as a template for both translation and RNA replication. However, a template that is actively being translated cannot function as a template for RNA replication, suggesting that there is a switch in template usage from translation to RNA replication. We demonstrate that the cleavage of PCBP2 by the poliovirus 3CD proteinase is a necessary step for efficient viral RNA replication and, as such, may be important for mediating a switch in template usage from translation to RNA replication. IMPORTANCE: Poliovirus, like all positive-strand RNA viruses that replicate in the cytoplasm of eukaryotic cells, uses its genomic RNA as a template for both viral protein synthesis and RNA replication. Given that these processes cannot occur simultaneously on the same template, poliovirus has evolved a mechanism(s) to facilitate the switch from using templates for translation to using them for RNA synthesis. This study explores one possible scenario for how the virus alters the functions of a host cell RNA binding protein to mediate, in part, this important transition.


Sujet(s)
Poliomyélite/métabolisme , Poliovirus/génétique , ARN viral/génétique , Protéines de liaison à l'ARN/métabolisme , Réplication virale , Protéases virales 3C , Cysteine endopeptidases/génétique , Cysteine endopeptidases/métabolisme , Régulation négative , Cellules HeLa , Humains , Poliomyélite/génétique , Poliomyélite/virologie , Poliovirus/enzymologie , Poliovirus/physiologie , Maturation post-traductionnelle des protéines , ARN viral/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines virales/génétique , Protéines virales/métabolisme
14.
Neuroscience ; 247: 309-18, 2013 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-23742846

RÉSUMÉ

Theiler's murine encephalomyelitis virus (TMEV) induces demyelination in susceptible strains of mice through a CD4(+) Th1 T cell-mediated immunopathological process. TMEV infection produces a syndrome in mice that resembles multiple sclerosis. In this work, we focused on the increased expression of the genes encoding voltage-gated Ca(2+) channel subunits in SJL/J mouse astrocytes infected in culture with a BeAn strain of TMEV. Affymetrix DNA murine genome U74v2 DNA microarray hybridized with cRNA from mock- and TMEV-infected astrocytes revealed the upregulation of four sequences encoding Ca(2+)-binding and Ca(2+) channel subunit proteins. The DNA hybridization results were further validated using conventional RT-PCR and quantitative RT-PCR, demonstrating the increased expression of mRNA encoding channel subunit proteins. Western blotting also showed the increased synthesis of L- and N-type channel subunit specific proteins after infection. The reduced expression and the functional upregulation of functional voltage-gated Ca(2+) channels in mock- and TMEV-infected cells, respectively, was demonstrated using voltage clamp experiments. TMEV infection in mouse astrocytes induced a Ca(2+) current with a density proportional to the amount of viral particles used for infection. The use of Ca(2+) channel blockers, nimodipine and ω-conotoxin-GVIA, showed that both functional L- and N-type Ca(2+) channels were upregulated in infected astrocytes. The upregulation of Ca(2+) channels in astrocytes after TMEV infection provides insight into the molecular processes and potential role of astrocyte Ca(2+) dysregulation in the pathophysiology of encephalomyelitis and is important for the development of novel therapeutic strategies leading to prevention of neurodegeneration.


Sujet(s)
Astrocytes/métabolisme , Astrocytes/virologie , Canaux calciques/biosynthèse , Poliomyélite/métabolisme , Theilovirus/pathogénicité , Régulation positive/physiologie , Animaux , Lignée cellulaire , Cellules cultivées , Cricetinae , Souris , Poliomyélite/physiopathologie , Poliomyélite/médecine vétérinaire
15.
PLoS Pathog ; 9(6): e1003401, 2013.
Article de Anglais | MEDLINE | ID: mdl-23762027

RÉSUMÉ

All positive strand (+RNA) viruses of eukaryotes replicate their genomes in association with membranes. The mechanisms of membrane remodeling in infected cells represent attractive targets for designing future therapeutics, but our understanding of this process is very limited. Elements of autophagy and/or the secretory pathway were proposed to be hijacked for building of picornavirus replication organelles. However, even closely related viruses differ significantly in their requirements for components of these pathways. We demonstrate here that infection with diverse picornaviruses rapidly activates import of long chain fatty acids. While in non-infected cells the imported fatty acids are channeled to lipid droplets, in infected cells the synthesis of neutral lipids is shut down and the fatty acids are utilized in highly up-regulated phosphatidylcholine synthesis. Thus the replication organelles are likely built from de novo synthesized membrane material, rather than from the remodeled pre-existing membranes. We show that activation of fatty acid import is linked to the up-regulation of cellular long chain acyl-CoA synthetase activity and identify the long chain acyl-CoA syntheatse3 (Acsl3) as a novel host factor required for polio replication. Poliovirus protein 2A is required to trigger the activation of import of fatty acids independent of its protease activity. Shift in fatty acid import preferences by infected cells results in synthesis of phosphatidylcholines different from those in uninfected cells, arguing that the viral replication organelles possess unique properties compared to the pre-existing membranes. Our data show how poliovirus can change the overall cellular membrane homeostasis by targeting one critical process. They explain earlier observations of increased phospholipid synthesis in infected cells and suggest a simple model of the structural development of the membranous scaffold of replication complexes of picorna-like viruses, that may be relevant for other (+)RNA viruses as well.


Sujet(s)
Coenzyme A ligases/biosynthèse , Acides gras/métabolisme , Régulation de l'expression des gènes codant pour des enzymes , Poliomyélite/enzymologie , Poliovirus/physiologie , Réplication virale/physiologie , Transport biologique actif , Cysteine endopeptidases/métabolisme , Cellules HeLa , Humains , Poliomyélite/génétique , Poliomyélite/métabolisme , Régulation positive , Protéines virales/métabolisme
16.
Mol Cell Biol ; 33(5): 1016-26, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23275440

RÉSUMÉ

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


Sujet(s)
Adenoviridae/physiologie , Hepacivirus/physiologie , Interactions hôte-pathogène , Poliovirus/physiologie , Protéines ribosomiques/métabolisme , Ribosomes/virologie , Infections à Adenoviridae/génétique , Infections à Adenoviridae/métabolisme , Infections à Adenoviridae/virologie , Lignée cellulaire , Techniques de knock-down de gènes , Cellules HeLa , Hépatite C/génétique , Hépatite C/métabolisme , Hépatite C/virologie , Humains , Poliomyélite/génétique , Poliomyélite/métabolisme , Poliomyélite/virologie , Biosynthèse des protéines , Protéines ribosomiques/génétique , Ribosomes/métabolisme , Réplication virale
17.
PLoS Pathog ; 8(11): e1003046, 2012.
Article de Anglais | MEDLINE | ID: mdl-23209416

RÉSUMÉ

The autophagic pathway acts as part of the immune response against a variety of pathogens. However, several pathogens subvert autophagic signaling to promote their own replication. In many cases it has been demonstrated that these pathogens inhibit or delay the degradative aspect of autophagy. Here, using poliovirus as a model virus, we report for the first time bona fide autophagic degradation occurring during infection with a virus whose replication is promoted by autophagy. We found that this degradation is not required to promote poliovirus replication. However, vesicular acidification, which in the case of autophagy precedes delivery of cargo to lysosomes, is required for normal levels of virus production. We show that blocking autophagosome formation inhibits viral RNA synthesis and subsequent steps in the virus cycle, while inhibiting vesicle acidification only inhibits the final maturation cleavage of virus particles. We suggest that particle assembly, genome encapsidation, and virion maturation may occur in a cellular compartment, and we propose the acidic mature autophagosome as a candidate vesicle. We discuss the implications of our findings in understanding the late stages of poliovirus replication, including the formation and maturation of virions and egress of infectious virus from cells.


Sujet(s)
Autophagie , Phagosomes/métabolisme , Poliomyélite/métabolisme , Poliovirus/physiologie , Virion/métabolisme , Assemblage viral/physiologie , Cellules HeLa , Humains , Concentration en ions d'hydrogène , Phagosomes/virologie , Réplication virale/physiologie
18.
J Virol ; 86(10): 5541-53, 2012 May.
Article de Anglais | MEDLINE | ID: mdl-22379090

RÉSUMÉ

Poliovirus (PV) modifies membrane-trafficking machinery in host cells for its viral RNA replication. To date, ARF1, ACBD3, BIG1/BIG2, GBF1, RTN3, and PI4KB have been identified as host factors of enterovirus (EV), including PV, involved in membrane traffic. In this study, we performed small interfering RNA (siRNA) screening targeting membrane-trafficking genes for host factors required for PV replication. We identified valosin-containing protein (VCP/p97) as a host factor of PV replication required after viral protein synthesis, and its ATPase activity was essential for PV replication. VCP colocalized with viral proteins 2BC/2C and 3AB/3B in PV-infected cells and showed an interaction with 2BC and 3AB but not with 2C and 3A. Knockdown of VCP did not suppress the replication of coxsackievirus B3 or Aichi virus. A VCP-knockdown-resistant PV mutant had an A4881G (a mutation of E253G in 2C) mutation, which is known as a determinant of a secretion inhibition-negative phenotype. However, knockdown of VCP did not affect the inhibition of cellular protein secretion caused by overexpression of each individual viral protein. These results suggested that VCP is a host factor required for viral RNA replication of PV among membrane-trafficking proteins and provides a novel link between cellular protein secretion and viral RNA replication.


Sujet(s)
Adenosine triphosphatases/métabolisme , Protéines du cycle cellulaire/métabolisme , Poliomyélite/métabolisme , Poliovirus/physiologie , Voie de sécrétion , Réplication virale , Adenosine triphosphatases/génétique , Protéines du cycle cellulaire/génétique , Lignée cellulaire , Humains , Poliomyélite/génétique , Poliomyélite/virologie , Poliovirus/génétique , Transport des protéines , Protéine contenant la valosine , Protéines virales/génétique , Protéines virales/métabolisme
19.
J Virol ; 86(1): 302-12, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-22072780

RÉSUMÉ

Replication of all positive-strand RNA viruses is intimately associated with membranes. Here we utilize electron tomography and other methods to investigate the remodeling of membranes in poliovirus-infected cells. We found that the viral replication structures previously described as "vesicles" are in fact convoluted, branching chambers with complex and dynamic morphology. They are likely to originate from cis-Golgi membranes and are represented during the early stages of infection by single-walled connecting and branching tubular compartments. These early viral organelles gradually transform into double-membrane structures by extension of membranous walls and/or collapsing of the luminal cavity of the single-membrane structures. As the double-membrane regions develop, they enclose cytoplasmic material. At this stage, a continuous membranous structure may have double- and single-walled membrane morphology at adjacent cross-sections. In the late stages of the replication cycle, the structures are represented mostly by double-membrane vesicles. Viral replication proteins, double-stranded RNA species, and actively replicating RNA are associated with both double- and single-membrane structures. However, the exponential phase of viral RNA synthesis occurs when single-membrane formations are predominant in the cell. It has been shown previously that replication complexes of some other positive-strand RNA viruses form on membrane invaginations, which result from negative membrane curvature. Our data show that the remodeling of cellular membranes in poliovirus-infected cells produces structures with positive curvature of membranes. Thus, it is likely that there is a fundamental divergence in the requirements for the supporting cellular membrane-shaping machinery among different groups of positive-strand RNA viruses.


Sujet(s)
Membranes intracellulaires/virologie , Poliomyélite/virologie , Poliovirus/physiologie , Réplication virale , Membrane cellulaire/métabolisme , Membrane cellulaire/virologie , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/virologie , Appareil de Golgi/métabolisme , Appareil de Golgi/virologie , Cellules HeLa , Humains , Membranes intracellulaires/métabolisme , Poliomyélite/métabolisme , Poliovirus/génétique , Protéines virales/génétique , Protéines virales/métabolisme
20.
J Virol ; 85(23): 12442-54, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21957303

RÉSUMÉ

In response to environmental stress and viral infection, mammalian cells form foci containing translationally silenced mRNPs termed stress granules (SGs). As aggregates of stalled initiation complexes, SGs are defined by the presence of translation initiation machinery in addition to mRNA binding proteins. Here, we report that cells infected with poliovirus (PV) can form SGs early that contain T-cell-restricted intracellular antigen 1 (TIA1), translation initiation factors, RNA binding proteins, and mRNA. However, this response is blocked as infection progresses, and a type of pseudo-stress granule remains at late times postinfection and contains TIA but lacks translation initiation factors, mRNA binding proteins, and most polyadenylated mRNA. This result was observed using multiple stressors, including viral infection, oxidative stress, heat shock, and endoplasmic reticulum stress. Multiple proteins required for efficient viral internal ribosome entry site-dependent translation are localized to SGs under stress conditions, providing a potential rationale for the evolution and maintenance of the SG inhibition phenotype. Further, the expression of a noncleavable form of the RasGAP-SH3 domain binding protein in PV-infected cells enables SGs whose constituents are consistent with the presence of stalled 48S translation preinitiation complexes to persist throughout infection. These results indicate that in poliovirus-infected cells, the functions of TIA self-aggregation and aggregation of stalled translation initiation complexes into stress granules are severed, leading to novel foci that contain TIA1 but lack other stress granule-defining components.


Sujet(s)
Granulations cytoplasmiques/métabolisme , Stress oxydatif , Poliomyélite/virologie , Poliovirus/pathogénicité , Protéines de liaison au poly(A)/métabolisme , ARN messager/physiologie , Technique de Western , Cytoplasme/métabolisme , Cytoplasme/virologie , Technique d'immunofluorescence , Cellules HeLa , Humains , Hybridation in situ , Facteurs initiation chaîne peptidique/métabolisme , Poliomyélite/génétique , Poliomyélite/métabolisme , Protéines de liaison au poly(A)/génétique , Biosynthèse des protéines , Sondes d'ARN , Protéines de liaison à l'ARN/métabolisme , Antigène intracellulaire-1 des lymphocytes T
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...