Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 9(10)2021 Oct 16.
Article in English | MEDLINE | ID: mdl-34696298

ABSTRACT

The development of a live-attenuated vaccine (LAV) for the prevention of human metapneumovirus (HMPV) infection is often hampered by the lack of highly efficient and scalable cell-based production systems that support eventual global vaccine production. Avian cell lines cultivated in suspension compete with traditional cell platforms used for viral vaccine manufacture. We investigated whether the DuckCelt®-T17 avian cell line (Vaxxel), previously described as an efficient production system for several influenza strains, could also be used to produce a new HMPV LAV candidate (Metavac®, SH gene-deleted A1/C-85473 HMPV). To that end, we characterized the operational parameters of MOI, cell density, and trypsin addition to achieve the optimal production of Metavac®, and demonstrated that the DuckCelt®-T17 cell line is permissive and well-adapted to the production of the wild-type A1/C-85473 HMPV and the Metavac® vaccine candidate. Moreover, our results confirmed that the LAV candidate produced in DuckCelt®-T17 cells conserves its advantageous replication properties in LLC-MK2 and 3D-reconstituted human airway epithelium models, and its capacity to induce efficient neutralizing antibodies in a BALB/c mouse model. Our results suggest that the DuckCelt®-T17 avian cell line is a very promising platform for the scalable in-suspension serum-free production of the HMPV-based LAV candidate Metavac®.

2.
J Infect Dis ; 223(6): 1052-1061, 2021 03 29.
Article in English | MEDLINE | ID: mdl-32726438

ABSTRACT

Human respiratory syncytial virus (HRSV) constitutes one the main causes of respiratory infection in neonates and infants worldwide. Transcriptome analysis of clinical samples using high-throughput technologies remains an important tool to better understand virus-host complex interactions in the real-life setting but also to identify new diagnosis/prognosis markers or therapeutics targets. A major challenge when exploiting clinical samples such as nasal swabs, washes, or bronchoalveolar lavages is the poor quantity and integrity of nucleic acids. In this study, we applied a tailored transcriptomics workflow to exploit nasal wash samples from children who tested positive for HRSV. Our analysis revealed a characteristic immune signature as a direct reflection of HRSV pathogenesis and highlighted putative biomarkers of interest such as IP-10, TMEM190, MCEMP1, and TIMM23.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Tract Infections , Child , Gene Expression Profiling , Humans , Infant , Infant, Newborn , Nasopharynx , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/immunology
3.
Microorganisms ; 8(12)2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33322535

ABSTRACT

An increasing amount of evidence indicates a relatively high prevalence of superinfections associated with coronavirus disease 2019 (COVID-19), including invasive aspergillosis, but the underlying mechanisms remain to be characterized. In the present study, to better understand the biological impact of superinfection, we determine and compare the host transcriptional response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) versus Aspergillus superinfection, using a model of reconstituted human airway epithelium. Our analyses reveal that both simple infection and superinfection induce strong deregulation of core components of innate immune and inflammatory responses, with a stronger response to superinfection in the bronchial epithelial model compared to its nasal counterpart. Our results also highlight unique transcriptional footprints of SARS-CoV-2 Aspergillus superinfection, such as an imbalanced type I/type III IFN, and an induction of several monocyte and neutrophil associated chemokines, that could be useful for the understanding of Aspergillus-associated COVID-19 and also the management of severe forms of aspergillosis in this specific context.

4.
Cell Rep Med ; 1(4): 100059, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32835306

ABSTRACT

In the current COVID-19 pandemic context, proposing and validating effective treatments represents a major challenge. However, the scarcity of biologically relevant pre-clinical models of SARS-CoV-2 infection imposes a significant barrier for scientific and medical progress, including the rapid transition of potentially effective treatments to the clinical setting. We use reconstituted human airway epithelia to isolate and then characterize the viral infection kinetics, tissue-level remodeling of the cellular ultrastructure, and transcriptional early immune signatures induced by SARS-CoV-2 in a physiologically relevant model. Our results emphasize distinctive transcriptional immune signatures between nasal and bronchial HAE, both in terms of kinetics and intensity, hence suggesting putative intrinsic differences in the early response to SARS-CoV-2 infection. Most important, we provide evidence in human-derived tissues on the antiviral efficacy of remdesivir monotherapy and explore the potential of the remdesivir-diltiazem combination as an option worthy of further investigation to respond to the still-unmet COVID-19 medical need.


Subject(s)
Antiviral Agents/pharmacology , Bronchi/virology , Nose/virology , Respiratory Mucosa/virology , SARS-CoV-2/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Airway Remodeling , Alanine/analogs & derivatives , Alanine/pharmacology , Animals , Bronchi/drug effects , Bronchi/immunology , Bronchi/ultrastructure , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Chlorocebus aethiops , Diltiazem/pharmacology , Drug Synergism , Humans , Immunity, Innate , Models, Biological , Nose/drug effects , Nose/immunology , Nose/ultrastructure , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/ultrastructure , SARS-CoV-2/growth & development , Vero Cells , COVID-19 Drug Treatment
5.
Antiviral Res ; 181: 104878, 2020 09.
Article in English | MEDLINE | ID: mdl-32679055

ABSTRACT

In response to the current pandemic caused by the novel SARS-CoV-2, identifying and validating effective therapeutic strategies is more than ever necessary. We evaluated the in vitro antiviral activities of a shortlist of compounds, known for their cellular broad-spectrum activities, together with drugs that are currently under evaluation in clinical trials for COVID-19 patients. We report the antiviral effect of remdesivir, lopinavir, chloroquine, umifenovir, berberine and cyclosporine A in Vero E6 cells model of SARS-CoV-2 infection, with estimated 50% inhibitory concentrations of 0.99, 5.2, 1.38, 3.5, 10.6 and 3 µM, respectively. Virus-directed plus host-directed drug combinations were also investigated. We report a strong antagonism between remdesivir and berberine, in contrast with remdesivir/diltiazem, for which we describe high levels of synergy, with mean Loewe synergy scores of 12 and peak values above 50. Combination of host-directed drugs with direct acting antivirals underscore further validation in more physiological models, yet they open up interesting avenues for the treatment of COVID-19.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Drug Repositioning , Pandemics , Pneumonia, Viral/drug therapy , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Animals , Berberine/pharmacology , COVID-19 , Chlorocebus aethiops , Chloroquine/pharmacology , Coronavirus Infections/virology , Cyclosporine/pharmacology , Drug Antagonism , Drug Combinations , Drug Synergism , Humans , Indoles/pharmacology , Lopinavir/pharmacology , Pneumonia, Viral/virology , SARS-CoV-2 , Vero Cells , COVID-19 Drug Treatment
7.
Vaccines (Basel) ; 7(4)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671656

ABSTRACT

Human metapneumovirus (HMPV) is a major pediatric respiratory pathogen with currently no specific treatment or licensed vaccine. Different strategies to prevent this infection have been evaluated, including live-attenuated vaccines (LAV) based on SH and/or G protein deletions. This approach showed promising outcomes but has not been evaluated further using different viral strains. In that regard, we previously showed that different HMPV strains harbor distinct in vitro fusogenic and in vivo pathogenic phenotypes, possibly influencing the selection of vaccine strains. In this study, we investigated the putative contribution of the low conserved SH or G accessory proteins in such strain-dependent phenotypes and generated recombinant wild type (WT) and SH- or G-deleted viruses derived from two different patient-derived HMPV strains, A1/C-85473 and B2/CAN98-75. The ΔSH and ΔG deletions led to different strain-specific phenotypes in both LLC-MK2 cell and reconstituted human airway epithelium models. More interestingly, the ΔG-85473 and especially ΔSH-C-85473 recombinant viruses conferred significant protection against HMPV challenge and induced immunogenicity against a heterologous strain. In conclusion, our results show that the viral genetic backbone should be considered in the design of live-attenuated HMPV vaccines, and that a SH-deleted virus based on the A1/C-85473 HMPV strain could be a promising LAV candidate as it is both attenuated and protective in mice while being efficiently produced in a cell-based system.

8.
Sci Rep ; 9(1): 11493, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31391513

ABSTRACT

Acute respiratory infections, a large part being of viral origin, constitute a major public health issue. To propose alternative and/or new therapeutic approaches, it is necessary to increase our knowledge about the interactions between respiratory viruses and their primary cellular targets using the most biologically relevant experimental models. In this study, we used RNAseq to characterize and compare the transcriptomic signature of infection induced by different major respiratory viruses (Influenza viruses, hRSV and hMPV) in a model of reconstituted human airway epithelia. Our results confirm the importance of several cellular pathways commonly or specifically induced by these respiratory viruses, such as the innate immune response or antiviral defense. A very interesting common feature revealed by the global virogenomic signature shared between hRSV, hMPV and influenza viruses is the global downregulation of cilium-related gene expression, in good agreement with experimental evaluation of mucociliary clearance. Beyond providing new information about respiratory virus/host interactions, our study also underlines the interest of using biologically relevant experimental models to study human respiratory viruses.


Subject(s)
Gene Expression Regulation/immunology , Host Microbial Interactions/genetics , Respiratory Mucosa/immunology , Respiratory Tract Infections/immunology , Transcriptome/immunology , Animals , Cell Culture Techniques/methods , Cell Line , Cilia/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Host Microbial Interactions/immunology , Humans , Immunity, Innate/genetics , Influenza, Human/immunology , Macaca mulatta , Metapneumovirus/immunology , RNA-Seq , Respiratory Mucosa/cytology , Respiratory Mucosa/virology , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Infections/virology , Signal Transduction/genetics , Signal Transduction/immunology
9.
PLoS Pathog ; 15(4): e1007689, 2019 04.
Article in English | MEDLINE | ID: mdl-30964929

ABSTRACT

NOD-like receptor protein 3 (NLRP3) inflammasome activation triggers caspase-1 activation-induced maturation of interleukin (IL)-1ß and IL-18 and therefore is important for the development of the host defense against various RNA viral diseases. However, the implication of this protein complex in human metapneumovirus (HMPV) disease has not been fully studied. Herein, we report that NLRP3 inflammasome plays a detrimental role during HMPV infection because NLRP3 inflammasome inhibition protected mice from mortality and reduced weight loss and inflammation without impacting viral replication. We also demonstrate that NLRP3 inflammasome exerts its deleterious effect via IL-1ß production since we observed reduced mortality, weight loss and inflammation in IL-1ß-deficient (IL-1ß-/-) mice, as compared to wild-type animals during HMPV infection. Moreover, the effect on these evaluated parameters was not different in IL-1ß-/- and wild-type mice treated with an NLRP3 inflammasome inhibitor. The production of IL-1ß was also abrogated in bone marrow derived macrophages deficient for NLRP3. Finally, we show that small hydrophobic protein-deleted recombinant HMPV (HMPV ΔSH) failed to activate caspase-1, which is responsible for IL-1ß cleavage and maturation. Furthermore, HMPV ΔSH-infected mice had less weight loss, showed no mortality and reduced inflammation, as compared to wild-type HMPV-infected mice. Thus, NLRP3 inflammasome activation seems to be triggered by HMPV SH protein in HMPV disease. In summary, once activated by the HMPV SH protein, NLRP3 inflammasome promotes the maturation of IL-1ß, which exacerbates HMPV-induced inflammation. Therefore, the blockade of IL-1ß production by using NLRP3 inflammasome inhibitors might be a novel potential strategy for the therapy and prevention of HMPV infection.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , Interleukin-1beta/physiology , Metapneumovirus/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Paramyxoviridae Infections/immunology , Retroviridae Proteins, Oncogenic/metabolism , Animals , Female , Humans , Inflammasomes/metabolism , Inflammation/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Paramyxoviridae Infections/virology , Recombinant Proteins/metabolism , Retroviridae Proteins, Oncogenic/immunology , Signal Transduction , Virus Replication
10.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30651364

ABSTRACT

Influenza A viruses (IAV) are known to modulate and "hijack" several cellular host mechanisms, including gene splicing and RNA maturation machineries. These modulations alter host cellular responses and enable an optimal expression of viral products throughout infection. The interplay between the host protein p53 and IAV, in particular through the viral nonstructural protein NS1, has been shown to be supportive for IAV replication. However, it remains unknown whether alternatively spliced isoforms of p53, known to modulate p53 transcriptional activity, are affected by IAV infection and contribute to IAV replication. Using a TP53 minigene, which mimics intron 9 alternative splicing, we have shown here that the NS1 protein of IAV changes the expression pattern of p53 isoforms. Our results demonstrate that CPSF4 (cellular protein cleavage and polyadenylation specificity factor 4) independently and the interaction between NS1 and CPSF4 modulate the alternative splicing of TP53 transcripts, which may result in the differential activation of p53-responsive genes. Finally, we report that CPSF4 and most likely beta and gamma spliced p53 isoforms affect both viral replication and IAV-associated type I interferon secretion. All together, our data show that cellular p53 and CPSF4 factors, both interacting with viral NS1, have a crucial role during IAV replication that allows IAV to interact with and alter the expression of alternatively spliced p53 isoforms in order to regulate the cellular innate response, especially via type I interferon secretion, and perform efficient viral replication.IMPORTANCE Influenza A viruses (IAV) constitute a major public health issue, causing illness and death in high-risk populations during seasonal epidemics or pandemics. IAV are known to modulate cellular pathways to promote their replication and avoid immune restriction via the targeting of several cellular proteins. One of these proteins, p53, is a master regulator involved in a large panel of biological processes, including cell cycle arrest, apoptosis, or senescence. This "cellular gatekeeper" is also involved in the control of viral infections, and viruses have developed a wide diversity of mechanisms to modulate/hijack p53 functions to achieve an optimal replication in their hosts. Our group and others have previously shown that p53 activity is finely modulated by different multilevel mechanisms during IAV infection. Here, we characterized IAV nonstructural protein NS1 and the cellular factor CPSF4 as major partners involved in the IAV-induced modulation of the TP53 alternative splicing that was associated with a strong modulation of p53 activity and notably the p53-mediated antiviral response.


Subject(s)
Influenza A virus/immunology , Influenza, Human/immunology , Tumor Suppressor Protein p53/immunology , Viral Nonstructural Proteins/immunology , mRNA Cleavage and Polyadenylation Factors/immunology , A549 Cells , Alternative Splicing/immunology , Cell Line, Tumor , Humans , Immunity, Innate/immunology , Influenza, Human/virology , Interferons/immunology , Virus Replication/immunology
11.
Sci Rep ; 8(1): 3746, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29487367

ABSTRACT

The interplay between influenza A viruses (IAV) and the p53 pathway has been reported in several studies, highlighting the antiviral contribution of p53. Here, we investigated the impact of IAV on the E3-ubiquitin ligase Mdm2, a major regulator of p53, and observed that IAV targets Mdm2, notably via its non-structural protein (NS1), therefore altering Mdm2 stability, p53/Mdm2 interaction and regulatory loop during the time-course of infection. This study also highlights a new antiviral facet of Mdm2 possibly increasing the list of its many p53-independent functions. Altogether, our work contributes to better understand the mechanisms underlining the complex interactions between IAV and the p53 pathway, for which both NS1 and Mdm2 arise as key players.


Subject(s)
Influenza A virus/physiology , Influenza, Human/metabolism , Influenza, Human/virology , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , Gene Expression , Gene Expression Regulation, Viral , Humans , Protein Stability , Protein Transport , Proto-Oncogene Proteins c-mdm2/genetics , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
12.
J Gen Virol ; 98(6): 1174-1180, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28613142

ABSTRACT

Human metapneumovirus (HMPV) is an important cause of respiratory tract infections. The mechanism by which its fusion (F) protein is responsible for variable cytopathic effects in vitro remains unknown. We aligned the F sequences of the poorly fusogenic B2/CAN98-75 strain and the hyperfusogenic A1/C-85473 strain and identified divergent residues located in the two functional heptad repeats domains (HRA and HRB). We generated recombinant viruses by inserting the mutations N135T-G139N-T143K-K166E-E167D in HRA and/or K479R-N482S in HRB, corresponding to swapped sequences from C-85473, into CAN98-75 background and investigated their impact on in vitro phenotype and fusogenicity. We demonstrated that the five HRA mutations enhanced the fusogenicity of the recombinant rCAN98-75 virus, almost restoring the phenotype of the wild-type rC-85473 strain, whereas HRB substitutions alone had no significant effect on cell-cell fusion. Altogether, our results support the importance of the HRA domain for an HMPV-triggered fusion mechanism and identify key residues that modulate syncytium formation.


Subject(s)
Cell Fusion , Giant Cells/virology , Metapneumovirus/growth & development , Mutant Proteins/metabolism , Mutation , Viral Fusion Proteins/metabolism , Animals , Cell Line , DNA Mutational Analysis , Epithelial Cells/physiology , Epithelial Cells/virology , Macaca mulatta , Metapneumovirus/genetics , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Conformation , Protein Domains , Recombination, Genetic , Reverse Genetics , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
13.
Sci Rep ; 6: 29006, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27373907

ABSTRACT

Influenza viruses replicate their single-stranded RNA genomes in the nucleus of infected cells and these replicated genomes (vRNPs) are then exported from the nucleus to the cytoplasm and plasma membrane before budding. To achieve this export, influenza viruses hijack the host cell export machinery. However, the complete mechanisms underlying this hijacking remain not fully understood. We have previously shown that influenza viruses induce a marked alteration of the nucleus during the time-course of infection and notably in the nucleolar compartment. In this study, we discovered that a major nucleolar component, called nucleolin, is required for an efficient export of vRNPs and viral replication. We have notably shown that nucleolin interacts with the viral nucleoprotein (NP) that mainly constitutes vRNPs. Our results suggest that this interaction could allow vRNPs to "catch" the host cell export machinery, a necessary step for viral replication.


Subject(s)
Influenza A Virus, H3N2 Subtype/physiology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Viral Core Proteins/metabolism , A549 Cells , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Dogs , Humans , Madin Darby Canine Kidney Cells , Nucleocapsid Proteins , Virus Replication , Nucleolin
14.
PLoS One ; 10(3): e0120283, 2015.
Article in English | MEDLINE | ID: mdl-25803584

ABSTRACT

Human metapneumovirus (HMPV) is an important cause of acute respiratory tract infections (ARTI) in children, elderly individuals and immunocompromised patients. In vitro, different HMPV strains can induce variable cytopathic effects ranging from large multinucleated syncytia to focal cell rounding. In this study, we investigated the impact of different in vitro phenotypes of two HMPV strains on viral replication and disease severity in a BALB/c mouse model. We first generated two recombinant GFP-expressing HMPV viruses: C-85473, a syncytium-inducing strain (rC-85473) belonging to the A1 subtype and CAN98-75, a focal cell rounding-inducing strain (rCAN98-75) of the B2 subtype. We subsequently exchanged the F genes of both strains to create the chimeric viruses rC-85473_F and rCAN98-75_F. We demonstrated that the F protein was the sole protein responsible for the syncytium phenotype and that viruses carrying a syncytium-inducing F protein replicated to significantly higher titers in vitro. In vivo, however, the virulence and replicative capacity of the different HMPV strains did not appear to be solely dependent on the F gene but also on the viral background, with the strains containing the C-85473 background inducing more weight loss as well as increased lung viral titers, pro-inflammatory cytokines and inflammation than strains containing the CAN98-75 background. In conclusion, the F protein is the main determinant of syncytium formation and replication kinetics in vitro, although it is not the only factor implicated in HMPV disease severity in mice.


Subject(s)
Giant Cells/pathology , Lung/pathology , Metapneumovirus/genetics , Metapneumovirus/pathogenicity , Paramyxoviridae Infections/pathology , Respiratory Tract Infections/pathology , Animals , Cell Line , Disease Models, Animal , Genes, Viral , Giant Cells/virology , Humans , Lung/virology , Metapneumovirus/physiology , Mice, Inbred BALB C , Paramyxoviridae Infections/virology , Respiratory Tract Infections/virology , Viral Proteins/genetics , Virus Replication
15.
mBio ; 5(3): e00070-14, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24825008

ABSTRACT

During their nuclear replication stage, influenza viruses hijack the host splicing machinery to process some of their RNA segments, the M and NS segments. In this review, we provide an overview of the current knowledge gathered on this interplay between influenza viruses and the cellular spliceosome, with a particular focus on influenza A viruses (IAV). These viruses have developed accurate regulation mechanisms to reassign the host spliceosome to alter host cellular expression and enable an optimal expression of specific spliced viral products throughout infection. Moreover, IAV segments undergoing splicing display high levels of similarity with human consensus splice sites and their viral transcripts show noteworthy secondary structures. Sequence alignments and consensus analyses, along with recently published studies, suggest both conservation and evolution of viral splice site sequences and structure for improved adaptation to the host. Altogether, these results emphasize the ability of IAV to be well adapted to the host's splicing machinery, and further investigations may contribute to a better understanding of splicing regulation with regard to viral replication, host range, and pathogenesis.


Subject(s)
Orthomyxoviridae/genetics , RNA Splicing , RNA, Viral , Animals , Gene Expression Regulation , Gene Expression Regulation, Viral , Humans , Orthomyxoviridae/metabolism , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/virology , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Nonstructural Proteins/genetics , Viral Tropism , Virus Replication
16.
FEBS Lett ; 587(18): 2965-71, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23954291

ABSTRACT

The interplay between influenza A viruses (IAV) and p53 has only been reported in a limited number of studies, mainly focusing on the antiviral role of p53. We investigated the impact of IAV infection on p53 stability and transcriptional activity. Our results indicate that IAV-induced stabilization of p53 only partially correlates with modulation of p53 transcriptional activity measured during infection. Moreover, we show that the viral non-structural protein 1 (NS1) is able to inhibit p53 transcriptional activity, in a promoter-dependent manner. Based on these data, we propose that NS1 may contribute to p53-mediated cell fate decision during IAV infection.


Subject(s)
Influenza A Virus, H3N2 Subtype/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Binding Sites , Cell Line , Dogs , Host-Pathogen Interactions , Humans , Influenza A Virus, H3N2 Subtype/genetics , Madin Darby Canine Kidney Cells , Promoter Regions, Genetic , Protein Binding , Protein Stability , Tumor Suppressor Protein p53/genetics , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...