Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Front Immunol ; 15: 1329846, 2024.
Article de Anglais | MEDLINE | ID: mdl-38529279

RÉSUMÉ

Understanding adaptive immunity against SARS-CoV-2 is a major requisite for the development of effective vaccines and treatments for COVID-19. CD4+ T cells play an integral role in this process primarily by generating antiviral cytokines and providing help to antibody-producing B cells. To empower detailed studies of SARS-CoV-2-specific CD4+ T cell responses in mouse models, we comprehensively mapped I-Ab-restricted epitopes for the spike and nucleocapsid proteins of the BA.1 variant of concern via IFNγ ELISpot assay. This was followed by the generation of corresponding peptide:MHCII tetramer reagents to directly stain epitope-specific T cells. Using this rigorous validation strategy, we identified 6 immunogenic epitopes in spike and 3 in nucleocapsid, all of which are conserved in the ancestral Wuhan strain. We also validated a previously identified epitope from Wuhan that is absent in BA.1. These epitopes and tetramers will be invaluable tools for SARS-CoV-2 antigen-specific CD4+ T cell studies in mice.


Sujet(s)
COVID-19 , SARS-CoV-2 , Animaux , Souris , Lymphocytes T CD4+ , Déterminants antigéniques des lymphocytes T , Nucléocapside/composition chimique , Peptides/composition chimique , SARS-CoV-2/composition chimique , Antigènes d'histocompatibilité de classe II/composition chimique , Glycoprotéine de spicule des coronavirus/composition chimique
2.
Nat Commun ; 15(1): 787, 2024 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-38278784

RÉSUMÉ

SARS-CoV-2-reactive T cells are detected in some healthy unexposed individuals. Human studies indicate these T cells could be elicited by the common cold coronavirus OC43. To directly test this assumption and define the role of OC43-elicited T cells that are cross-reactive with SARS-CoV-2, we develop a model of sequential infections with OC43 followed by SARS-CoV-2 in HLA-B*0702 and HLA-DRB1*0101 Ifnar1-/- transgenic mice. We find that OC43 infection can elicit polyfunctional CD8+ and CD4+ effector T cells that cross-react with SARS-CoV-2 peptides. Furthermore, pre-exposure to OC43 reduces subsequent SARS-CoV-2 infection and disease in the lung for a short-term in HLA-DRB1*0101 Ifnar1-/- transgenic mice, and a longer-term in HLA-B*0702 Ifnar1-/- transgenic mice. Depletion of CD4+ T cells in HLA-DRB1*0101 Ifnar1-/- transgenic mice with prior OC43 exposure results in increased viral burden in the lung but no change in virus-induced lung damage following infection with SARS-CoV-2 (versus CD4+ T cell-sufficient mice), demonstrating that the OC43-elicited SARS-CoV-2 cross-reactive T cell-mediated cross-protection against SARS-CoV-2 is partially dependent on CD4+ T cells. These findings contribute to our understanding of the origin of pre-existing SARS-CoV-2-reactive T cells and their effects on SARS-CoV-2 clinical outcomes, and also carry implications for development of broadly protective betacoronavirus vaccines.


Sujet(s)
COVID-19 , Coronavirus humain OC43 , Humains , Souris , Animaux , SARS-CoV-2 , Souris transgéniques , Chaines HLA-DRB1/génétique , Lymphocytes T CD4+ , Glycoprotéine de spicule des coronavirus
3.
bioRxiv ; 2023 Nov 17.
Article de Anglais | MEDLINE | ID: mdl-38014059

RÉSUMÉ

Understanding adaptive immunity against SARS-CoV-2 is a major requisite for the development of effective vaccines and treatments for COVID-19. CD4+ T cells play an integral role in this process primarily by generating antiviral cytokines and providing help to antibody-producing B cells. To empower detailed studies of SARS-CoV-2-specific CD4+ T cell responses in mouse models, we comprehensively mapped I-Ab-restricted epitopes for the spike and nucleocapsid proteins of the BA.1 variant of concern via IFNγ ELISpot assay. This was followed by the generation of corresponding peptide:MHCII tetramer reagents to directly stain epitope-specific T cells. Using this rigorous validation strategy, we identified 6 reliably immunogenic epitopes in spike and 3 in nucleocapsid, all of which are conserved in the ancestral Wuhan strain. We also validated a previously identified epitope from Wuhan that is absent in BA.1. These epitopes and tetramers will be invaluable tools for SARS-CoV-2 antigen-specific CD4+ T cell studies in mice.

4.
Sci Adv ; 8(29): eabn1440, 2022 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-35857835

RÉSUMÉ

Measles virus, Nipah virus, and multiple other paramyxoviruses cause disease outbreaks in humans and animals worldwide. The paramyxovirus matrix (M) protein mediates virion assembly and budding from host cell membranes. M is thus a key target for antivirals, but few high-resolution structures of paramyxovirus M are available, and we lack the clear understanding of how viral M proteins interact with membrane lipids to mediate viral assembly and egress that is needed to guide antiviral design. Here, we reveal that M proteins associate with phosphatidylserine and phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] at the plasma membrane. Using x-ray crystallography, electron microscopy, and molecular dynamics, we demonstrate that PI(4,5)P2 binding induces conformational and electrostatic changes in the M protein surface that trigger membrane deformation, matrix layer polymerization, and virion assembly.

5.
Cell Rep ; 39(2): 110655, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35417697

RÉSUMÉ

Zika virus (ZIKV) and dengue virus (DENV) are arthropod-borne pathogenic flaviviruses that co-circulate in many countries. To understand some of the pressures that influence ZIKV evolution, we mimic the natural transmission cycle by repeating serial passaging of ZIKV through cultured mosquito cells and either DENV-naive or DENV-immune mice. Compared with wild-type ZIKV, the strains passaged under both conditions exhibit increased pathogenesis in DENV-immune mice. Application of reverse genetics identifies an isoleucine-to-valine mutation (I39V) in the NS2B proteins of both passaged strains that confers enhanced fitness and escape from pre-existing DENV immunity. Introduction of I39V or I39T, a naturally occurring homologous mutation detected in recent ZIKV isolates, increases the replication of wild-type ZIKV in human neuronal precursor cells and laboratory-raised mosquitoes. Our data indicate that ZIKV strains with enhanced transmissibility and pathogenicity can emerge in DENV-naive or -immune settings, and that NS2B-I39 mutants may represent ZIKV variants of interest.


Sujet(s)
Virus de la dengue , Dengue , Infection par le virus Zika , Virus Zika , Animaux , Anticorps antiviraux , Réactions croisées , Virus de la dengue/génétique , Souris , Mutation/génétique , Virus Zika/génétique
6.
Curr Biol ; 31(15): 3440-3449.e7, 2021 08 09.
Article de Anglais | MEDLINE | ID: mdl-34146484

RÉSUMÉ

Macroautophagy (hereafter referred to as autophagy) is a conserved process that promotes cellular homeostasis through the degradation of cytosolic components, also known as cargo. During autophagy, cargo is sequestered into double-membrane vesicles called autophagosomes, which are predominantly transported in the retrograde direction to the perinuclear region to fuse with lysosomes, thus ensuring cargo degradation.1 The mechanisms regulating directional autophagosomal transport remain unclear. The ATG8 family of proteins associates with autophagosome membranes2 and plays key roles in autophagy, including the movement of autophagosomes. This is achieved via the association of ATG8 with adaptor proteins like FYCO1, involved in the anterograde transport of autophagosomes toward the cell periphery.1,3-5 We previously reported that phosphorylation of LC3B/ATG8 on threonine 50 (LC3B-T50) by the Hippo kinase STK4/MST1 is required for autophagy through unknown mechanisms.6 Here, we show that STK4-mediated phosphorylation of LC3B-T50 reduces the binding of FYCO1 to LC3B. In turn, impairment of LC3B-T50 phosphorylation decreases starvation-induced perinuclear positioning of autophagosomes as well as their colocalization with lysosomes. Moreover, a significantly higher number of LC3B-T50A-positive autophagosomes undergo aberrant anterograde movement to axonal tips in mammalian neurons and toward the periphery of mammalian cells. Our data support a role of a nutrient-sensitive STK4-LC3B-FYCO1 axis in the regulation of the directional transport of autophagosomes, a key step of the autophagy process, via the post-translational modification of LC3B.


Sujet(s)
Autophagosomes , Protéines associées aux microtubules , Maturation post-traductionnelle des protéines , Animaux , Autophagosomes/métabolisme , Autophagie , Famille de la protéine-8 associée à l'autophagie/génétique , Famille de la protéine-8 associée à l'autophagie/métabolisme , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Phosphorylation
7.
Cell Rep ; 35(2): 108986, 2021 04 13.
Article de Anglais | MEDLINE | ID: mdl-33852858

RÉSUMÉ

The Ebola virus matrix protein VP40 forms distinct structures linked to distinct functions in the virus life cycle. Dimeric VP40 is a structural protein associated with virus assembly, while octameric, ring-shaped VP40 is associated with transcriptional control. In this study, we show that suitable nucleic acid is sufficient to trigger a dynamic transformation of VP40 dimer into the octameric ring. Deep sequencing reveals a binding preference of the VP40 ring for the 3' untranslated region of cellular mRNA and a guanine- and adenine-rich binding motif. Complementary analyses of the nucleic-acid-induced VP40 ring by native mass spectrometry, electron microscopy, and X-ray crystal structures at 1.8 and 1.4 Å resolution reveal the stoichiometry of RNA binding, as well as an interface involving a key guanine nucleotide. The host factor-induced structural transformation of protein structure in response to specific RNA triggers in the Ebola virus life cycle presents unique opportunities for therapeutic inhibition.


Sujet(s)
Régions 3' non traduites , Ebolavirus/génétique , Guanine/composition chimique , Interactions hôte-pathogène/génétique , Nucléoprotéines/composition chimique , Protéines du core viral/composition chimique , Sites de fixation , Cristallographie aux rayons X , Ebolavirus/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme , Expression des gènes , Guanine/métabolisme , Cellules HEK293 , Séquençage nucléotidique à haut débit , Humains , Modèles moléculaires , Nucléoprotéines/génétique , Nucléoprotéines/métabolisme , Motifs nucléotidiques , Liaison aux protéines , Structure en hélice alpha , Structure en brin bêta , Motifs et domaines d'intéraction protéique , Multimérisation de protéines , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Protéines du core viral/génétique , Protéines du core viral/métabolisme , Assemblage viral/génétique , Libération de particules virales/génétique
8.
Trends Microbiol ; 28(8): 605-618, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32507543

RÉSUMÉ

SARS-Coronavirus-2 (SARS-CoV-2) causes Coronavirus disease 2019 (COVID-19), an infectious respiratory disease causing thousands of deaths and overwhelming public health systems. The international spread of SARS-CoV-2 is associated with the ease of global travel, and societal dynamics, immunologic naiveté of the host population, and muted innate immune responses. Based on these factors and the expanding geographic scale of the disease, the World Health Organization (WHO) declared the COVID-19 outbreak a pandemic-the first caused by a coronavirus. In this review, we summarize the current epidemiological status of COVID-19 and consider the virological and immunological lessons, animal models, and tools developed in response to prior SARS-CoV and MERS-CoV outbreaks that can serve as resources for development of SARS-CoV-2 therapeutics and vaccines. In particular, we discuss structural insights into the SARS-CoV-2 spike protein, a major determinant of transmissibility, and discuss key molecular aspects that will aid in understanding and fighting this new global threat.


Sujet(s)
Betacoronavirus/composition chimique , Betacoronavirus/immunologie , Infections à coronavirus/épidémiologie , Infections à coronavirus/virologie , Pandémies , Pneumopathie virale/épidémiologie , Pneumopathie virale/virologie , Animaux , COVID-19 , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/thérapie , Modèles animaux de maladie humaine , Humains , Pandémies/prévention et contrôle , Pneumopathie virale/prévention et contrôle , Pneumopathie virale/thérapie , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/immunologie
9.
mBio ; 10(4)2019 07 23.
Article de Anglais | MEDLINE | ID: mdl-31337716

RÉSUMÉ

The filoviruses are etiological agents of life-threatening hemorrhagic fever with high mortality rate and risk of potential outbreak. Among members of this family, the Ebola (EBOV), Sudan (SUDV), and Marburg (MARV) viruses are considered the most pathogenic for humans. The ebolavirus nucleoprotein (NP) is the most abundant protein in infected cells and is essential for viral transcription and replication; thus, it represents an attractive target for therapeutic intervention. Here, we present the structure of SUDV NP in complex with the amino-terminal portion of the phosphoprotein VP35 at 2.3 Å. This structure captures VP35 chaperoning SUDV NP in a monomeric and RNA-free state. This transient state has been proposed to be key to maintaining a pool of monomeric and RNA-free NPs prior to NP-NP polymerization and encapsidation of the viral RNA genome. This structure also reveals a newly visualized interaction between NP and VP35, a well-defined beta sheet that is not present in previous structures. Affinity binding assays demonstrate that this beta sheet is essential for maintaining the high-affinity interaction between VP35 and a hydrophobic pocket on SUDV NP, and electron microscopy indicates the importance of this binding interaction to the oligomeric state and assembly of NP in human cells. Complementary structure-directed mutagenesis identifies critical residues conserved across the filovirus family that could be targeted by broadly effective antivirals.IMPORTANCE Outbreaks of the filoviruses can be unpredictable in timing, location, and identity of the causative virus, with each of Ebola virus, Sudan virus, Bundibugyo virus, and Marburg virus reemerging in the last several years to cause human disease with 30 to 90% lethality. The 2014-2016 outbreak in particular, with nearly 30,000 patients, highlighted the ability of these viruses to emerge unexpectedly and spread rapidly. Two ebolavirus outbreaks have emerged this year, yet we still lack FDA-approved drugs with pan-filovirus activity to treat existing and emergent ebolaviruses. For all filoviruses, the interaction between the nucleoprotein and the phosphoprotein is essential for the virus life cycle and is a potential target for therapeutic intervention. In this report, we describe the crystal structure of the SUDV nucleoprotein with the interacting domain of the viral phosphoprotein, and we identify residues critical for high-affinity interaction and for control of the oligomeric state of the nucleoprotein. Structural comparison of this heterodimer with other members of the filovirus family allowed us to find conserved and essential atomic features that will facilitate understanding of the virus life cycle and the rational design of antivirals.


Sujet(s)
Ebolavirus/effets des médicaments et des substances chimiques , Filoviridae/effets des médicaments et des substances chimiques , Nucléoprotéines/composition chimique , Protéines virales régulatrices ou accessoires/composition chimique , Cristallographie aux rayons X , Filoviridae/pathogénicité , Phosphoprotéines/composition chimique , Structure en brin bêta , Motifs et domaines d'intéraction protéique
10.
Sci Rep ; 8(1): 1230, 2018 01 19.
Article de Anglais | MEDLINE | ID: mdl-29352168

RÉSUMÉ

Influenza virus stablishes a network of virus-host functional interactions, which depends on chromatin dynamic and therefore on epigenetic modifications. Using an unbiased search, we analyzed the epigenetic changes at DNA methylation and post-translational histone modification levels induced by the infection. DNA methylation was unaltered, while we found a general decrease on histone acetylation, which correlates with transcriptional inactivation and may cooperate with the impairment of cellular transcription that causes influenza virus infection. A particular increase in H3K79 methylation was observed and the use of an inhibitor of the specific H3K79 methylase, Dot1L enzyme, or its silencing, increased influenza virus replication. The antiviral response was reduced in conditions of Dot1L downregulation, since decreased nuclear translocation of NF-kB complex, and IFN-ß, Mx1 and ISG56 expression was detected. The data suggested a control of antiviral signaling by methylation of H3K79 and consequently, influenza virus replication was unaffected in IFN pathway-compromised, Dot1L-inhibited cells. H3K79 methylation also controlled replication of another potent interferon-inducing virus such as vesicular stomatitis virus, but did not modify amplification of respiratory syncytial virus that poorly induces interferon signaling. Epigenetic methylation of H3K79 might have an important role in controlling interferon-induced signaling against viral pathogens.


Sujet(s)
Méthylation de l'ADN , Épigenèse génétique , Code histone , Interactions hôte-pathogène , Orthomyxoviridae/physiologie , Protéines adaptatrices de la transduction du signal , Animaux , Lignée cellulaire tumorale , Chiens , Cellules HEK293 , Histone-lysine N-methyltransferase , Humains , Interféron bêta/métabolisme , Cellules rénales canines Madin-Darby , Methyltransferases/génétique , Methyltransferases/métabolisme , Protéines de résistance aux myxovirus/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéines de liaison à l'ARN , Facteurs de transcription/métabolisme , Réplication virale
12.
mBio ; 7(2): e00085-16, 2016 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-27094326

RÉSUMÉ

UNLABELLED: Influenza A viruses generate annual epidemics and occasional pandemics of respiratory disease with important consequences for human health and the economy. Therefore, a large effort has been devoted to the development of new anti-influenza virus drugs directed to viral targets, as well as to the identification of cellular targets amenable to anti-influenza virus therapy. Here we have addressed the identification of such potential cellular targets by screening collections of drugs approved for human use. We reasoned that screening with a green fluorescent protein-based recombinant replicon system would identify cellular targets involved in virus transcription/replication and/or gene expression and hence address an early stage of virus infection. By using such a strategy, we identified Montelukast (MK) as an inhibitor of virus multiplication. MK inhibited virus gene expression but did not alter viral RNA synthesis in vitro or viral RNA accumulation in vivo The low selectivity index of MK prevented its use as an antiviral, but it was sufficient to identify a new cellular pathway suitable for anti-influenza virus intervention. By deep sequencing of RNA isolated from mock- and virus-infected human cells, treated with MK or left untreated, we showed that it stimulates the PERK-mediated unfolded protein stress response. The phosphorylation of PERK was partly inhibited in virus-infected cells but stimulated in MK-treated cells. Accordingly, pharmacological inhibition of PERK phosphorylation led to increased viral gene expression, while inhibition of PERK phosphatase reduced viral protein synthesis. These results suggest the PERK-mediated unfolded protein response as a potential cellular target to modulate influenza virus infection. IMPORTANCE: Influenza A viruses are responsible for annual epidemics and occasional pandemics with important consequences for human health and the economy. The unfolded protein response is a defense mechanism fired by cells when the demand of protein synthesis and folding is excessive, for instance, during an acute virus infection. In this report, we show that influenza virus downregulates the unfolded protein response mediated by the PERK sensor, while Montelukast, a drug used to treat asthma in humans, specifically stimulated this response and downregulated viral protein synthesis and multiplication. Accordingly, we show that PERK phosphorylation was reduced in virus-infected cells and increased in cells treated with Montelukast. Hence, our studies suggest that modulation of the PERK-mediated unfolded protein response is a target for influenza virus inhibition.


Sujet(s)
Acétates/pharmacologie , Virus de la grippe A/effets des médicaments et des substances chimiques , Grippe humaine/enzymologie , Quinoléines/pharmacologie , Réponse aux protéines mal repliées , eIF-2 Kinase/métabolisme , Cyclopropanes , Humains , Virus de la grippe A/génétique , Virus de la grippe A/physiologie , Grippe humaine/génétique , Grippe humaine/physiopathologie , Grippe humaine/virologie , Phosphorylation , Sulfures , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Protéines virales/génétique , Protéines virales/métabolisme , Réplication virale/effets des médicaments et des substances chimiques , eIF-2 Kinase/génétique
13.
Sci Rep ; 6: 20744, 2016 Feb 11.
Article de Anglais | MEDLINE | ID: mdl-26864902

RÉSUMÉ

The influenza A virus polymerase associates with a number of cellular transcription-related factors, including the RNA polymerase II (RNAP II). We previously described that the cellular protein hCLE/C14orf166 interacts with and stimulates influenza virus polymerase as well as RNAP II activities. Here we show that, despite the considerable cellular shut-off observed in infected cells, which includes RNAP II degradation, hCLE protein levels increase throughout infection in a virus replication-dependent manner. Human and avian influenza viruses of various subtypes increase hCLE levels, but other RNA or DNA viruses do not. hCLE colocalises and interacts with viral ribonucleoproteins (vRNP) in the nucleus, as well as in the cytoplasm late in infection. Furthermore, biochemical analysis of purified virus particles and immunoelectron microscopy of infected cells show hCLE in virions, in close association with viral vRNP. These findings indicate that hCLE, a cellular protein important for viral replication, is one of the very few examples of transcription factors that are incorporated into particles of an RNA-containing virus.


Sujet(s)
Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/génétique , Ribonucléoprotéines/génétique , Transactivateurs/génétique , Protéines virales/génétique , Virion/génétique , Cellules A549 , Animaux , Noyau de la cellule/métabolisme , Noyau de la cellule/ultrastructure , Noyau de la cellule/virologie , Cytoplasme/métabolisme , Cytoplasme/ultrastructure , Cytoplasme/virologie , Chiens , Régulation de l'expression des gènes , Cellules HEK293 , Interactions hôte-pathogène , Humains , Sous-type H1N1 du virus de la grippe A/métabolisme , Sous-type H1N1 du virus de la grippe A/ultrastructure , Sous-type H3N2 du virus de la grippe A/métabolisme , Sous-type H3N2 du virus de la grippe A/ultrastructure , Sous-type H9N2 du virus de la grippe A/métabolisme , Sous-type H9N2 du virus de la grippe A/ultrastructure , Cellules rénales canines Madin-Darby , Microscopie immunoélectronique , Protéolyse , RNA polymerase II/génétique , RNA polymerase II/métabolisme , Ribonucléoprotéines/métabolisme , Transactivateurs/métabolisme , Protéines virales/métabolisme , Virion/métabolisme , Virion/ultrastructure , Réplication virale
14.
Virus Res ; 212: 78-84, 2016 Jan 02.
Article de Anglais | MEDLINE | ID: mdl-26321158

RÉSUMÉ

Influenza A viruses generate annual epidemics and occasional pandemics of respiratory disease with important consequences for human health and economy. To establish a productive infection, influenza viruses interact with cellular factors to favour their own replication and to suppress antiviral cell responses. Although most virus-host interaction studies have been centred on cell protein factors, most of the human transcriptome comprises non-coding RNAs, as miRNAs and lncRNAs. The latter are key cellular regulators in many cellular processes, including transcriptional and post-transcriptional regulation. Influenza virus infection induces the differential expression of hundreds of potential lncRNAs, some of which are related to the antiviral pathways activated by the cell while others may be deregulated by the infection to allow efficient virus multiplication. Although our knowledge on the role of cellular lncRNAs for influenza virus replication and pathogenesis is still at its infancy, several lncRNAs have been described to influence the cell innate response to the virus by altering the histone modification at specific sites, by interaction with specific transcription factors or directly stimulating in cis the expression of specific IFN-induced genes. In addition, at least one lncRNA appears to be required for virus multiplication in an IFN-independent way.


Sujet(s)
Virus de la grippe A/physiologie , Grippe humaine/métabolisme , Grippe humaine/virologie , ARN long non codant/métabolisme , Animaux , Régulation de l'expression des gènes viraux , Interactions hôte-pathogène , Humains , Virus de la grippe A/génétique , Grippe humaine/génétique , ARN long non codant/génétique , Protéines virales/génétique , Protéines virales/métabolisme
15.
J Virol ; 89(19): 10023-30, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26202233

RÉSUMÉ

UNLABELLED: Transcription and replication of influenza A virus are carried out in the nuclei of infected cells in the context of viral ribonucleoproteins (RNPs). The viral polymerase responsible for these processes is a protein complex composed of the PB1, PB2, and PA proteins. We previously identified a set of polymerase-associated cellular proteins by proteomic analysis of polymerase-containing intracellular complexes expressed and purified from human cells. Here we characterize the role of NXP2/MORC3 in the infection cycle. NXP2/MORC3 is a member of the Microrchidia (MORC) family that is associated with the nuclear matrix and has RNA-binding activity. Influenza virus infection led to a slight increase in NXP2/MORC3 expression and its partial relocalization to the cytoplasm. Coimmunoprecipitation and immunofluorescence experiments indicated an association of NXP2/MORC3 with the viral polymerase and RNPs during infection. Downregulation of NXP2/MORC3 by use of two independent short hairpin RNAs (shRNAs) reduced virus titers in low-multiplicity infections. Consistent with these findings, analysis of virus-specific RNA in high-multiplicity infections indicated a reduction of viral RNA (vRNA) and mRNA after NXP2/MORC3 downregulation. Silencing of NXP2/MORC3 in a recombinant minireplicon system in which virus transcription and replication are uncoupled showed reductions in cat mRNA and chloramphenicol acetyltransferase (CAT) protein accumulation but no alterations in cat vRNA levels, suggesting that NXP2/MORC3 is important for influenza virus transcription. IMPORTANCE: Influenza virus infections appear as yearly epidemics and occasional pandemics of respiratory disease, with high morbidity and occasional mortality. Influenza viruses are intracellular parasites that replicate and transcribe their genomic ribonucleoproteins in the nuclei of infected cells, in a complex interplay with host cell factors. Here we characterized the role of the human NXP2/MORC3 protein, a member of the Microrchidia family that is associated with the nuclear matrix, during virus infection. NXP2/MORC3 associates with the viral ribonucleoproteins in infected cells. Downregulation of NXP2/MORC3 reduced virus titers and accumulations of viral genomic RNA and mRNAs. Silencing of NXP2/MORC3 in an influenza virus CAT minireplicon system diminished CAT protein and cat mRNA levels but not genomic RNA levels. We propose that NXP2/MORC3 plays a role in influenza virus transcription.


Sujet(s)
Adenosine triphosphatases/physiologie , Protéines de liaison à l'ADN/physiologie , Virus de la grippe A/physiologie , Virus de la grippe A/pathogénicité , Réplication virale/physiologie , Adenosine triphosphatases/antagonistes et inhibiteurs , Adenosine triphosphatases/génétique , Lignée cellulaire , Chloramphenicol O-acetyltransferase/génétique , Chloramphenicol O-acetyltransferase/métabolisme , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/génétique , Techniques de knock-down de gènes , Cellules HEK293 , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/physiologie , Humains , Sous-type H1N1 du virus de la grippe A , Sous-type H3N2 du virus de la grippe A , Virus de la grippe A/génétique , Protéomique , ARN messager/génétique , ARN messager/métabolisme , ARN viral/génétique , ARN viral/métabolisme , Transcription génétique , Protéines virales/génétique , Protéines virales/métabolisme , Réplication virale/génétique
16.
PLoS Pathog ; 7(11): e1002397, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-22114566

RÉSUMÉ

The influenza A virus RNA polymerase is a heterotrimeric complex responsible for viral genome transcription and replication in the nucleus of infected cells. We recently carried out a proteomic analysis of purified polymerase expressed in human cells and identified a number of polymerase-associated cellular proteins. Here we characterise the role of one such host factors, SFPQ/PSF, during virus infection. Down-regulation of SFPQ/PSF by silencing with two independent siRNAs reduced the virus yield by 2-5 log in low-multiplicity infections, while the replication of unrelated viruses as VSV or Adenovirus was almost unaffected. As the SFPQ/PSF protein is frequently associated to NonO/p54, we tested the potential implication of the latter in influenza virus replication. However, down-regulation of NonO/p54 by silencing with two independent siRNAs did not affect virus yields. Down-regulation of SFPQ/PSF by siRNA silencing led to a reduction and delay of influenza virus gene expression. Immunofluorescence analyses showed a good correlation between SFPQ/PSF and NP levels in infected cells. Analysis of virus RNA accumulation in silenced cells showed that production of mRNA, cRNA and vRNA is reduced by more than 5-fold but splicing is not affected. Likewise, the accumulation of viral mRNA in cicloheximide-treated cells was reduced by 3-fold. In contrast, down-regulation of SFPQ/PSF in a recombinant virus replicon system indicated that, while the accumulation of viral mRNA is reduced by 5-fold, vRNA levels are slightly increased. In vitro transcription of recombinant RNPs generated in SFPQ/PSF-silenced cells indicated a 4-5-fold reduction in polyadenylation but no alteration in cap snatching. These results indicate that SFPQ/PSF is a host factor essential for influenza virus transcription that increases the efficiency of viral mRNA polyadenylation and open the possibility to develop new antivirals targeting the accumulation of primary transcripts, a very early step during infection.


Sujet(s)
Virus de la grippe A/physiologie , Épissage des ARN , Réplication virale/génétique , Lignée cellulaire tumorale , Régulation négative , Cellules HEK293 , Humains , Virus de la grippe A/génétique , Cinétique , Facteur d'épissage associé à PTB , Polyadénylation , Interférence par ARN , ARN viral/métabolisme , Protéines de liaison à l'ARN , Ribonucléoprotéines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE