Cellular transcriptional profiling in human lung epithelial cells infected by different subtypes of influenza A viruses reveals an overall down-regulation of the host p53 pathway
11 pages
English

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris

Cellular transcriptional profiling in human lung epithelial cells infected by different subtypes of influenza A viruses reveals an overall down-regulation of the host p53 pathway

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus
11 pages
English
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus

Description

Influenza viruses can modulate and hijack several cellular signalling pathways to efficiently support their replication. We recently investigated and compared the cellular gene expression profiles of human lung A549 cells infected by five different subtypes of human and avian influenza viruses (Josset et al. Plos One 2010). Using these transcriptomic data, we have focused our analysis on the modulation of the p53 pathway in response to influenza infection. Results Our results were supported by both RT-qPCR and western blot analyses and reveal multiple alterations of the p53 pathway during infection. A down-regulation of mRNA expression was observed for the main regulators of p53 protein stability during infection by the complete set of viruses tested, and a significant decrease in p53 mRNA expression was also observed in H5N1 infected cells. In addition, several p53 target genes were also down-regulated by these influenza viruses and the expression of their product reduced. Conclusions Our data reveal that influenza viruses cause an overall down-regulation of the host p53 pathway and highlight this pathway and p53 protein itself as important viral targets in the altering of apoptotic processes and in cell-cycle regulation.

Informations

Publié par
Publié le 01 janvier 2011
Nombre de lectures 4
Langue English
Poids de l'ouvrage 2 Mo

Extrait

Terrier et al. Virology Journal 2011, 8:285
http://www.virologyj.com/content/8/1/285
RESEARCH Open Access
Cellular transcriptional profiling in human lung
epithelial cells infected by different subtypes
of influenza A viruses reveals an overall
down-regulation of the host p53 pathway
1,2† 1,3† 4 2 1 1Olivier Terrier , Laurence Josset , Julien Textoris , Virginie Marcel , Gaëlle Cartet , Olivier Ferraris ,
4 1,3 5 2 1*Catherine N’Guyen , Bruno Lina , Jean-Jacques Diaz , Jean-Christophe Bourdon and Manuel Rosa-Calatrava
Abstract
Background: Influenza viruses can modulate and hijack several cellular signalling pathways to efficiently support
their replication. We recently investigated and compared the cellular gene expression profiles of human lung A549
cells infected by five different subtypes of human and avian influenza viruses (Josset et al. Plos One 2010). Using
these transcriptomic data, we have focused our analysis on the modulation of the p53 pathway in response to
influenza infection.
Results: Our results were supported by both RT-qPCR and western blot analyses and reveal multiple alterations of
the p53 pathway during infection. A down-regulation of mRNA expression was observed for the main regulators of
p53 protein stability during infection by the complete set of viruses tested, and a significant decrease in p53 mRNA
expression was also observed in H5N1 infected cells. In addition, several p53 target genes were also
downregulated by these influenza viruses and the expression of their product reduced.
Conclusions: Our data reveal that influenza viruses cause an overall down-regulation of the host p53 pathway and
highlight this pathway and p53 protein itself as important viral targets in the altering of apoptotic processes and in
cell-cycle regulation.
Background viruses [3]. Current human circulating influenza A
Influenza viruses belong to the Orthomyxoviridae family subtypes are H1N1 and H3N2. While extensive viral
of enveloped viruses containing a segmented genome of diversity is responsible for the subtype-specific virus-host
single stranded negative RNA. Among the three influ- interactions, many common functional features are also
enza types (A, B and C), type A is the most virulent shared among viruses. Influenza infection alters host
celpathogen with a diversity represented by the combina- lular homeostasis via the combination of the
virallytion of 16 H and 9 N different subtypes (e.g. H1N1, induced alteration of biological machineries/pathways and
H5N1) [1]. Influenza A viruses are the most serious the cellular antiviral response triggered by intracellular
threat to public health, with the potential to cause glo- signalling cascades. Influenza viruses are able to activate/
bal pandemics as was illustrated in 2009 with the emer- inhibit and hijack several cellular signalling pathways to
efficiently support their own replication [4].gence of H1N1 SOIV [2].
All known subtypes of the influenza A virus are main- The development of high-throughput ‘omic’ studies
tained in wild waterfowl, the natural reservoir of these has increased our understanding of viral-host
interactions. Numerous studies have described host gene
expression modifications induced upon viral infection* Correspondence: manuel.rosa-calatrava@univ-lyon1.fr
† Contributed equally in vitro, in animal models and in patients [5-9]. For
exam1Laboratoire de Virologie et Pathologie Humaine VirPath, Université Claude
ple,aglobalmRNAprofilingstudyofMDCK-infected
Bernard Lyon 1, Université de Lyon, Lyon, France
cells has revealed an important role of the NF-kappaBFull list of author information is available at the end of the article
© 2011 Terrier et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in
any medium, provided the original work is properly cited.Terrier et al. Virology Journal 2011, 8:285 Page 2 of 11
http://www.virologyj.com/content/8/1/285
signalling pathway for the H5N1 subtype and to a lesser described in our previous study [17]. Confluent A549 cells
extent for the currently circulating H3N2 strain [10]. were infected with influenza viruses at a MOI of 1 or 10-3
Other intracellular signalling cascades are also induced by for one hour in a minimal volume of DMEM
supplemeninfection, in particular the mitogen-activated protein ted with 2 mM L-glutamine, 100 U of penicillin/mL,
kinase (MAPK) and the PI3K/Akt pathways, both of which 100μg of streptomycin sulphate/mL and 0.5μg of trypsin/
activate downstream transcription factors thereby affecting mL (infection medium) at 37°C. The cells were then
overhost gene expression [11,12]. While a small number of laid with fresh infection medium and incubated at 37°C
transcriptional studies have compared the cellular for 24 h. Viral kinetics for the six different influenza
viruses have been determined previously [15]. No markersresponse induced by infection with highly pathogenic
strains such as H1N1 1918 and H5N1 to that induced of apoptosis were detected at 24h post infection (MOI 1)
with low pathogenic strains, a systematic analysis and by western blot (capspase 3 cleavage) and no signs of cell
comparison of host cell mRNA expression during infec- death were observed (data not shown).
tion by several genetically diverging influenza subtypes has
not yet been performed [7,8,13,14]. Microarray analysis
Using a nylon microarray, we recently characterized Gene expression data of A549 cells infected by H1N1,
common gene expression changes induced during the H3N2, H5N1, H5N2 or H7N1 influenza viruses or
infection of human lung epithelial cells by five different mock infected (five replicates in each group) were
influenza A viruses. This shared signature was exploited obtained in our previous study [15]. Data sets are
availto find new molecules that act on host metabolic path- able publicly from the Gene Expression Omnibus (GEO)
ways to bring about an antiviral effect against several database (http://www.ncbi.nlm.nih.gov/geo/) under
subtypes [15]. Moreover, our systematic transcriptomic accession number GSE22319. Five supervised analyses
study also provided a gene expression database with between groups of infected versus mock samples were
which we compared the cellular responses induced by conducted using the Significance Analysis of Microarray
different influenza viruses. In the present study, we used algorithm (SAM) [18], and the siggenes library (v1.18.0)
the same data set to analyze strain specificity in regard [19]. Ingenuity Pathway Analysis 5.0 (IPA) (Ingenuity
to several host pathways. We observed that each viral Systems, Redwood City, CA, USA) was used to select,
signature was mainly associated with cellular pathways annotate, and visualize genes according to function and
linked to cell death and cell cycle progression (i.e. cell pathway (Gene Ontology). Additional gene annotation
growth). At the crossroads of all of these cellular pro- was provided by the Interferome Database [20] and
cesses, the p53 pathway became the focus of our investi- DAVID database (http://david.abcc.ncifcrf.gov/). Heat
gation. Our analyses, validated by both RT-qPCR and maps were produced by the heat map function from R
western blot analyses, reveal multiple alterations within that uses hierarchical clustering with Euclidean distance
the p53 pathway during influenza A infection. metric and the complete linkage method to generate the
hierarchical tree (http://www.r-project.org/).
Methods
Cell lines, viruses and infection Validation by RT-qPCR
Human lung epithelial A549 cells (ATCC CCL-185) or Total RNAs were extracted using the RNAeasy Mini Kit
human colon carcinoma HCT116 cell lines were grown as (Qiagen). Reverse-transcription was performed on 1 μg
monolayers in Dulbecco’s modified Eagle’smedium of total RNAs using the Superscript II enzyme
(Invitro(DMEM, Gibco), supplemented with 10% heat-inactivated gen) at 42°C. Quantification of the level of different
foetal bovine serum, 2 mM L-glutamine, 100 U/mL of mRNAs of interest was performed by real-time PCR on
penicillin and 100 mg/mL of streptomycin sulphate, at a MX3005P apparatus (Stratagene). Briefly, 20 ng of
37°C. The HCT116 cell lines containing a p53 wild-type cDNAs were amplified using 0.8 μM of each primer,
(p53+/+) and a p53-deleted derivative (p53-/-) were 0.4 μM of probe (cf. Table 1) and 1X Taqman Universal
gifts from Dr. Bert Vogelstein (John Hopkins University, Master Mix (Applied Biosystems). All data were
normalBaltimore, MD, USA) [16]. Influenza viruses A/New Cale- ized to the internal standard Actin. For each single-well
donia/20/99 (H1N1), A/Moscow/10/99 (H3N2), A/Lyon/ amplification reaction, a threshold cycle (Ct) was
969/09 (H1N1 SOIV), A/Turkey/582/2006 (H5N1), observed in the exponential phase of amplification.
RelaA/Finch/England/2051/94 (H5N2), and A/Chicken/Italy/ tive changes in gene expression were determined using
2076/99 (H7N1) were produced in MDCK cells in EMEM the 2ΔΔCt method and reported as the n-fold difference
supplemented with 2 mM L-glutamine, 100 U/mL of peni- relative to a control cDNA (mock, uninfected cells)
precillin, 100 mg/mL of streptomycin sulphate and 1 mg/mL pared in parallel with the experimental cDNAs (in

  • Univers Univers
  • Ebooks Ebooks
  • Livres audio Livres audio
  • Presse Presse
  • Podcasts Podcasts
  • BD BD
  • Documents Documents