Abstract As a highly pathogenic influenza virus, H5N1 avian influenza virus (AIV) poses a great threat to poultry production and public health. H5N1 AIV has a small genome and, therefore, relies heavily on its host cellular machinery to replicate. To develop a comprehensive understanding of how H5N1 AIV rewires host cellular machinery during the course of infection, it is crucial to identify which host proteins and complexes come into physical contact with the viral proteins. Here, we utilized affinity purification mass spectrometry (AP-MS) to systematically determine the physical interactions of 11 H5N1 AIV proteins with host proteins in chicken DF1 cells. We identified with high confidence 1,043 H5N1 AIV–chicken interactions involving 621 individual chicken proteins and uncovered a number of host proteins and complexes that were targeted by the viral proteins. Specifically, we revealed that chicken Staufen double-stranded RNA-binding protein 2 interacts with AIV non-structural protein 1 (NS1) and promotes the replication of the virus by enhancing the nuclear export of NS1 mRNA. This dataset facilitates a more comprehensive and detailed understanding of how the host machinery is manipulated during the course of H5N1 AIV infection. Keywords: H5N1 AIV, AP-MS, Stau2, NS1, chicken Introduction Influenza A virus (IAV) is a segmented, single-stranded, negative-sense RNA virus that has adapted to infect multiple species. This virus causes annual epidemics and recurring pandemics, which have huge impacts on public health. IAV particles have two viral surface glycoproteins (hemagglutinin, HA; neuraminidase, NA) and one matrix-2 protein (M2). Inside the virion, all eight viral RNA (vRNA) segments bind three RNA polymerases (polymerase acid protein, PA; polymerase basic protein 1, PB1; and 2, PB2) and are encapsidated by the nucleoprotein (NP) to form the viral ribonucleoprotein (vRNP) complexes ([33]1–[34]5). The vRNPs are surrounded by a layer of the matrix protein, M1, which lines the envelope ([35]6). Upon infection by influenza, the host cells detect the viral RNA through pathogen sensors, and the major gene products of the influenza virus mediate the viral life cycle and modulate cellular processes ([36]7). Viruses rely on host cellular functions to replicate, and thus, they hijack the host cell machinery and rewire it for their own needs. Several proteomic studies have used affinity purification mass spectrometry approaches to identify a series of cellular factors that interact with IAV proteins ([37]8–[38]10). However, knowledge of common and strain-specific interactions remains incomplete, and how these interactions control host defense and viral infection remains to be fully elucidated. A comprehensive understanding of host–virus interactions would greatly improve our understanding of the viral life cycle and host resistance mechanisms. Here, we applied the AP-MS technology to uncover a wide array of host proteins, complexes, and pathways that are hijacked by H5N1 avian influenza virus (AIV) during the course of infection. We constructed an H5N1 AIV–chicken protein interaction map, and in addition to replicating previously identified host factors, we uncovered several novel interactions. Among the novel factors, chicken Staufen double-stranded RNA-binding protein 2 (STAU2) was found to be a crucial component when the viral mRNA is transported during the replication stage of the viral life cycle. Importantly, STAU2 interacts with the influenza NS1 protein and promotes the replication of H5N1 AIV by promoting the transport of NS1 mRNA from the nucleus to the cytoplasm. Materials and Methods Cells and Virus Chicken embryonic fibroblast (DF1) cells, Madin–Darby canine kidney (MDCK) cells, and human embryonic kidney cells (293T) were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (Gibco), 100 μg/ml streptomycin, and 100 U/ml penicillin at 37°C under a humidified atmosphere of 5% CO[2]. The highly pathogenic H5N1 strain A/mallard/Huadong/S/2005 (SY) ([39]11) was propagated in 10-day-old specific pathogen-free embryonic chicken eggs. The H5N1 influenza virus strain 178 (GenBank Accession No. [40]AY737296-737300) was isolated from a chicken in Guangdong, China, by the MOA Key Laboratory for Animal Vaccine Development, P.R. China. The experiments that involved live viruses were performed in a biosafety cabinet with HEPA filters in a biosafety level 3 laboratory in Yangzhou University or South China Agricultural University. Plasmid Construction The H5N1 AIV genes were amplified by high-fidelity DNA polymerase (TransGen), and cDNA derived from the H5N1 virus (A/Chicken/ShanXi/2/2006) was used as the template. To construct the FLAG-tagged C-terminus fusion proteins, a 3 × FLAG tag was inserted into the C-terminus of the pcDNA 3.1 vector, and the PA, PB1, PB2, NP, HA, NA, M1, and NS1 genes were cloned upstream of the tag using the Seamless Assembly Cloning Kit (CloneSmarter). For the GFP-tagged proteins, a GFP tag was inserted into the C-terminus of the pcDNA 3.1 vector, and the NS2, M2, and PB1-F2 genes were cloned upstream of the GFP tag using the same method. All the expression vectors were validated by sequencing. Antibodies The following antibodies were used in this study: anti-FLAG [Abmart, # M20008L, WB (1:2,000)], anti-Myc [Abmart, # M20002L, WB (1:2,000)], and anti-Lamin B1 [Abcam, # ab16048, WB (1:2,000)]. Protein Co-immunoprecipitation and Western Blotting The transiently transfected cells were washed twice with phosphate-buffered saline (PBS) and were then lysed in radioimmunoprecipitation assay (RIPA) buffer (50 mM Tris–HCl, pH 7.4, 150 mM NaCl, 0.25% deoxycholic acid, 1% NP-40, 1 mM EDTA, and 0.5% SDS supplemented with protease inhibitor, Roche). The whole-cell lysate was firstly precleared with a protein A/G slurry (Millipore) and was then incubated with 40 μl of the anti-Flag affinity gel (Sigma-Aldrich) at 4°C for 2 h or with 10 μl of the GFP antibody at 4°C overnight, and then, it was incubated with 40 μl of the protein A/G slurry (Millipore) at 4°C for 2 h. The immunoprecipitated samples were washed four times with RIPA buffer and twice with 54K buffer (50 mM Tris–HCl, pH 7.4, 150 mM NaCl, and 0.25% Triton X-100 supplemented with protease inhibitor). The FLAG tag-associated proteins were eluted with 250 ng/μl of the Flag peptide (Sigma) by rocking the samples on a tilted rotator at 4°C for 2 h. The GFP tag-associated proteins were eluted with ammonium hydroxide at 4°C for 2 h, and the supernatant was collected with a vacuum centrifugal concentrator. For Western blotting, SDS electrophoresis was performed, and the separated proteins were transferred to polyvinylidene fluoride (PVDF) membranes. The membranes were blocked and then incubated with the corresponding antibodies. The proteins were visualized using the Immobilon Western Chemiluminescent HRP Substrate (Millipore). Mass Spectrometry Strict experimental controls were used for the MS analysis. The immunoprecipitated samples from the empty FLAG-transfected cells (empty FLAG control)/empty GFP-transfected cells (empty GFP control) and the protein complexes that were pulled down by the normal IgG (IgG control) were also subjected to mass spectrometry for identification. All the proteins identified in these two sets of controls were excluded from consideration as H5N1 AIV interacting proteins. The authentic FLAG-precipitated proteins and the GFP-precipitated proteins associated with H5N1 AIV were examined in triplicate. Proteins that were enriched by co-immunoprecipitation were separated by SDS-PAGE, and the entire lane was cut and sent for tryptic digestion. Proteins pulled down by the anti-FLAG beads or the anti-GFP antibody were digested with trypsin for 20 h at room temperature. The peptides were extracted twice with 50% aqueous acetonitrile containing 0.1% formic acid, dried in a SpeedVac, and then desalted using Sep-Pak C18 cartridges. Tandem mass tag (TMT) reagents (Thermo Fisher) were used to label the purified peptides according to the manufacturer's instructions. Briefly, the TMT labeling reagents, in anhydrous acetonitrile, were carefully added to the desalted peptides and incubated for 1 h at room temperature, after which the reactions were stopped by hydroxylamine, and the TMT-labeled peptides separated by reverse phase (RP) chromatography. The first dimension RP separation by micro-liquid chromatography (LC) was performed on an Ultimate 3000 System (Thermo Fisher) using an Xbridge C18 RP column (5 μm, 150 Å, 250 mm × 4.6 mm i.d., Waters). The samples were reconstituted in 5–10 ml of mobile phase A (2% acetonitrile, 0.1% formic acid, pH adjusted to 10.0 with NH[4]OH). The peptides were monitored at 214 nm, and 1-min fractions were collected, dried, and reconstituted in 20 μl of 0.1% (v/v) formic acid in water for the nano-LC-MS/MS analyses, in which the fractions were further separated on a C18 column (75 μm inner diameter, 150 mm length) with a flow rate of 250 nl/min. A gradient was formed, and the peptides were eluted with increasing concentrations of solvent B (98% acetonitrile, 0.1% formic acid, pH to 10.0, as above). This second-dimension separation was performed on an Orbitrap Q Exactive mass spectrometer that was operated in data-dependent acquisition mode using Xcalibur 3.0 software. The scan range was from m/z 300 to 1,800, with a resolution of 70,000 at m/z 400. A full scan followed by 20 data-dependent MS/MS scans was acquired with collision-induced dissociation having a normalized collision energy of 35%. The MS/MS spectra obtained from each LC-MS/MS run were searched against a protein database using the Proteome Discoverer searching algorithm. The precursor ion mass tolerance was set at 20 ppm, and the fragment ion mass tolerance was 20 mmu. One missed cleavage by trypsin was allowed. Oxidation (Met) was chosen as the variable modification, and carbamidomethyl (Cys) and TMT6plex were chosen as the fixed modifications. Significance Analysis of Interactome The MS data were analyzed using the Significance Analysis of Interactome (SAINT) tool, and only the results with a SAINT score >0.9 were considered for the subsequent analysis. To define the high-probability interaction sets, we selected the interactions at a probability threshold of 0.9, which was approximately equivalent to an estimated false discovery rate (FDR) of 2% ([41]12). Gene Ontology Term Enrichment and Pathway Enrichment Analysis Gene Ontology (GO) enrichment and pathway enrichment were conducted using the Database for Annotation, Visualization and Integrated Discovery (DAVID) ([42]13). Domain Enrichment Analysis The protein domain enrichment analysis was conducted for the H5N1 AIV-associated host proteins using FunRich ([43]http://funrich.org/index.html). Immunofluorescence Staining and Confocal Microscopy Analysis Chicken DF1 cells transiently transfected with FLAG-NS1 and Myc-STAU2 plasmids were cultured for 24 h and were then fixed in 4% paraformaldehyde for 30 min at room temperature and were permeabilized with 0.1% Triton X-100 (Sigma, # T8787) for 15 min. FLAG-NS1 cells were incubated with an anti-FLAG antibody (Abmart, # M20008L) and Myc-STAU2 cells with an anti-Myc antibody (Abmart, # M20002L), both at a 1:1,000 dilution for 2 h. After three washes in PBS, the cells were subsequently incubated with a FITC-conjugated secondary antibody (Abcam, # ab6785) for FLAG and with a Cy3.5-conjugated secondary antibody (Abcam, # ab6954) for Myc, both at a 1:1,000 dilution for 1 h. The nuclei were stained with 6-diamidino-2-phenylindole (DAPI) (Sigma, # D9542, 1:500). Finally, the cells were visualized with a confocal microscope (Nikon A1 R MP). RNA Interference All the small interfering RNAs (siRNAs) used in this study were designed and synthesized by Guangzhou Ruibo (Guangzhou, China). Chicken DF1 cells, at 90% confluence in six-well plates, were transfected with 100 nM of an effective siRNA specific for the chicken STAU2 gene (Gene ID: [44]420184: siSTAU2, sense 5′-CCTACAAGCTCTCCAGAAT-3′; siNC, sense 5′-GUGAACGAACUCCUUAAUUTT-3′). Human 293T cells, at 65% confluence in six-well plates, were transfected with 100 nM of an effective siRNA specific for the human STAU2 gene (Gene ID: [45]27067: siSTAU2, sense 5′-CCAAGGGAUGAACCCUAUUTT-3′). All the siRNAs were transfected into the cells using Lipofectamine 3000 (Life Technologies). Cell Culture and Viral Transfections The highly pathogenic H5N1 strain A/mallard/Huadong/S/2005 (YS), showing a high virulence in mice, was isolated from a mallard. Chicken DF1 embryonic fibroblast cells, 293T cells, and MDCK cells were cultured in DMEM supplemented with 10% fetal bovine serum (Gibco), 100 μg/ml streptomycin, and 100 units/ml penicillin at 37°C under a humidified atmosphere of 5% CO[2]. The transfection with siRNA or plasmid was performed with Lipofectamine 3000 (Life Technologies). The cells were harvested for protein extraction 48 h after the transfection. Virus Titration The 50% tissue culture infectious dose assay (TCID[50]) was used to evaluate progeny virus production. The siRNA-STAU2- or siRNA-NC-transfected DF1 cells grown in six-well plates were infected with H5N1 AIV, A/mallard/Huadong/S/2005 (SY), at a multiplicity of infection (MOI) of 0.1. After a 1-h incubation, the supernatants were discarded, and the cells were washed twice with PBS. Subsequently, 2 ml of DMEM without fetal bovine serum was added. The supernatants were collected at 12 and 18 h postinfection (h.p.i.), and the virus titers were determined in the MDCK cells. The virus titer was calculated as the TCID[50] per 0.1 ml using the Reed and Muench method. The data are shown as the means ± standard deviations (SD) from three independent experiments. An independent-sample t-test was used to analyze the TCID[50] results. For all the tests, P ≤ 0.05 was considered significant. Extraction of Nuclear and Cytoplasmic RNA The DF1 cells were transfected with an effective siRNA specific for STAU2 for 12 h and then were transfected with the NS1-FLAG plasmid for 24 h. The DF1 cells and 293T cells were infected with the H5N1 avian influenza virus for 10 h. The fresh cells were collected and were then separated into the nuclear and cytoplasmic fractions to extract the nuclear RNA and cytoplasmic RNA, respectively, using the PARIS™ Kit (Life Technologies, USA). Real-Time PCR Total cellular RNA was extracted by TRIzol (Invitrogen) according to the manufacturer's instructions. Next, cDNA was generated from 1 μg of RNA using the PrimeScript RT reagent kit with gDNA Eraser (TaKaRa). The target mRNAs were quantified by real-time PCR using SYBR Green Master Mix. The data were normalized to the expression of the housekeeping genes GAPDH and β-actin. NS1 expression in the nucleus was normalized to the expression of U6, which is only expressed in the nucleus. The sequences of the PCR primers used to amplify target genes are listed as follows: STAU2-chicken: sense 5′-AGTGCCTAAAATTTTCTATGT-3′, antisense 5′-GCTTCCCCATTCTGAGGTAAT-3′; STAU2-human: sense 5′-AAGCACTGCAGAATGAA-3′, antisense 5′-GCAATTTCAAACACTAA-3′; NS1: sense 5′-TGCGGGAAAGCAGATAGT-3′, antisense 5′-TGGGCATGAGCATGAACC-3′; GAPDH-chicken: sense 5′-CCCCCATGTTTGTGATGGGT-3′, antisense 5′-TGATGGCATGGACAGTGGTC-3′; GAPDH-human: sense 5′-AACCATGAGAAGTATGAC-3′, antisense 5′-GATGGCATGGACTGTGGT-3′; β-actin-chicken: sense 5′-GAGAAATTGTGCGTGACATCA-3′, antisense 5′-CCTGAACCTCTCATTGCCA-3′; β-actin-human: sense 5′-CTCCATCCTGGCCTCGCTGT-3′, antisense 5′-GCTGTCACCTTCACCGTTCC-3′; U6-chicken: sense 5′-CTCGCTTCGGCAGCACATAT-3′, antisense 5′-TGGAACGCTTCACGAATTTG-3′; and U6-human: sense 5′-TTCGGCAGCACATATA-3′, antisense 5′-ATATGGAACGCTTCAC-3′. Results Identification of Host Proteins Associated With 11 Viral H5N1 AIV Proteins We aimed to systematically and quantitatively identify host proteins associated with H5N1 AIV proteins using AP-MS. To this end, eight FLAG-tagged viral proteins (i.e., PA, PB1, PB2, NP, HA, NA, M1, and NS1) and three GFP-tagged viral proteins (i.e., PB1-F2, M2, and NS2) of an avian influenza virus (A/Chicken/ShanXi/2/2006, H5N1 subtype) were individually expressed in chicken DF1 cells. Affinity purification was carried out using FLAG tag or GFP tag antibodies to immunoprecipitate the host proteins associated with the tagged viral proteins. The Western blot results showed that all the viral genes were expressed in the chicken cells ([46]Supplementary Figure 1). The immunoprecipitated samples from the plasmid-transfected cells were separated by SDS-PAGE for MS analysis ([47]Figure 1A). In order to generate AP-MS data with a high degree of confidence, all the samples (including those from the empty FLAG/GFP-transfected cells and the IgG control samples) were subjected to the SAINT analysis ([48]12), and only the results with a SAINT score >0.9 were considered for the subsequent analysis. The MS analysis of the coprecipitated proteins identified 621 host proteins in total, and among these, 103, 41, 23, 131, 81, 213, 151, 38, 17, 186, and 62 coprecipitated with the viral PA ([49]14), PB1, PB1-F2, PB2, NP, HA, NA, M1, M2, NS1, and NS2 proteins, respectively ([50]Figure 1B, [51]Supplementary Table 1). Figure 1. [52]Figure 1 [53]Open in a new tab Affinity purification of H5N1 avian influenza virus (AIV) proteins. (A) Flowchart of the proteomic affinity purification mass spectrometry (AP-MS) method used to define the H5N1 AIV–host interactome. (B) The number of host proteins identified by AP-MS that interact with individual H5N1 avian influenza virus proteins. H5N1 AIV–Chicken Interactome We next plotted a network representation of the 621 H5N1 AIV–chicken protein interactions identified in this study ([54]Figure 2A). This network contained nodes corresponding to 11 H5N1 AIV proteins (purple) and the 621 host proteins derived from the chicken DF1 cells (pink and green). The outermost pink nodes and the innermost green nodes represent the host proteins that interact with only one viral protein or with multiple viral proteins, respectively. The network structure suggests that a large proportion of the interacting host proteins are not only factors for this particular influenza virus but may have more general biological functions and interact with multiple viral proteins. Figure 2. [55]Figure 2 [56]Open in a new tab Global landscape of the H5N1 AIV–chicken protein complexes. In total, 1,043 H5N1 AIV–chicken interactions are represented, linking 11 H5N1 AIV proteins and 621 chicken factors. (A) Types of host interacting proteins. Chicken proteins that interact with H5N1 AIV proteins (purple nodes in the middle ring) are denoted with two colors: the pink nodes in the outer ring represent the host proteins that interact with only one specific virus protein, while the green nodes in the inner ring represent the host proteins that interact with a variety of viral proteins. (B) Network representation of the H5N1 AIV–human PPIs. The host–pathogen interaction map for H5N1 AIV contained 1,475 interactions between 11 influenza virus proteins (red nodes) and 621 cellular proteins (blue and pink nodes). Some of the host proteins interact with the virus in the form of protein complexes (pink nodes). Next, we analyzed all of the H5N1 AIV–host protein interactions using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) ([57]15) and plotted a network representation ([58]Figure 2B, [59]Supplementary Table 2) incorporating both the MS and STRING analyses ([60]Supplementary Table 3) using Cytoscape. In this network, the red nodes represent the viral proteins, the remaining are the host interaction proteins, and the pink nodes form the subnetworks in the protein–protein interaction (PPI) map. The green lines represent the interactions among the virus and host proteins, while the gray lines represent interactions among the host proteins. In addition to interactions with the viral proteins, it was evident that many of the host interacting proteins also interacted with the other host proteins, and some even associated with the viral proteins in the form of protein complexes ([61]Figure 2B). GO, Pathway, and Domain Enrichment Analysis of the Host Interacting Partners of H5N1 AIV We performed a GO analysis and pathway enrichment on the host interactors of the H5N1 AIV proteins using the DAVID analysis tool. A number of crucial GO terms and pathways were identified as enriched in the AIV–chicken interacting proteins ([62]Tables 1, [63]2), indicating that these signaling pathways and associated genes play important roles in the life cycle of the influenza virus. Table 1. Gene Ontology terms enriched among host proteins interacting with H5N1 AIV. GO term Count Involved genes/total genes (%) P-value Molecular function Poly(A) RNA binding 116 22.4 1.30E-37 ATP binding 106 20.5 2.50E-15 RNA binding 37 7.1 1.10E-11 Nucleotide binding 35 6.8 4.40E-10 ATP-dependent RNA helicase activity 13 2.5 4.80E-07 Actin filament binding 13 2.5 1.10E-06 Structural constituent of ribosome 21 4.1 2.30E-05 mRNA binding 14 2.7 2.70E-05 Translation initiation factor activity 10 1.9 5.20E-05 Double-stranded RNA binding 9 1.7 3.30E-04 Cellular component Extracellular exosome 149 28.8 2.90E-22 Membrane 86 16.6 1.40E-19 Myelin sheath 29 5.6 2.30E-13 Nucleoplasm 89 17.2 7.40E-11 Focal adhesion 39 7.5 1.10E-10 Catalytic step 2 spliceosome 16 3.1 5.60E-08 Mitochondrial inner membrane 23 4.4 4.10E-07 U1 snRNP 7 1.4 3.90E-06 Nucleolus 40 7.7 7.50E-06 U5 snRNP 7 1.4 1.30E-05 U2 snRNP 7 1.4 1.30E-05 Biological process RNA secondary structure unwinding 10 1.9 3.90E-06 Translation 20 3.9 1.50E-05 Protein folding 15 2.9 2.80E-05 IRES-dependent viral translational initiation 5 1 3.00E-05 mRNA splicing, via spliceosome 11 2.1 2.20E-04 Negative regulation of translation 8 1.5 3.60E-04 Osteoblast differentiation 11 2.1 4.00E-04 Viral translational termination-reinitiation 4 0.8 4.80E-04 mRNA processing 10 1.9 7.40E-04 Regulation of translational initiation 6 1.2 8.50E-04 Protein import into nucleus 8 1.5 1.00E-03 [64]Open in a new tab Table 2. Pathway enrichments of host proteins interacting with H5N1 AIV. Term Count Involved genes/total genes (%) P-value Spliceosome 28 5.4 8.40E-11 Proteasome 11 2.1 4.40E-05 RNA transport 22 4.2 4.50E-05 Citrate cycle (TCA cycle) 9 1.7 2.10E-04 Ribosome 19 3.7 3.00E-04 DNA replication 9 1.7 3.60E-04 Carbon metabolism 16 3.1 7.40E-04 Protein processing in endoplasmic reticulum 20 3.9 1.50E-03 Pyruvate metabolism 8 1.5 4.20E-03 Salmonella infection 11 2.1 7.00E-03 Biosynthesis of antibiotics 21 4.1 8.50E-03 Aminoacyl-tRNA biosynthesis 8 1.5 1.40E-02 Ribosome biogenesis in eukaryotes 10 1.9 2.00E-02 Mismatch repair 5 1 3.20E-02 Pentose phosphate pathway 5 1 4.40E-02 [65]Open in a new tab Protein domains provide insight into the preferential structure of an interaction partner. To survey the binding preferences of the H5N1 AIV