Free access
Research Article
15 September 2014

Influenza A and B Virus Intertypic Reassortment through Compatible Viral Packaging Signals

ABSTRACT

Influenza A and B viruses cocirculate in humans and together cause disease and seasonal epidemics. These two types of influenza viruses are evolutionarily divergent, and exchange of genetic segments inside coinfected cells occurs frequently within types but never between influenza A and B viruses. Possible mechanisms inhibiting the intertypic reassortment of genetic segments could be due to incompatible protein functions of segment homologs, a lack of processing of heterotypic segments by influenza virus RNA-dependent RNA polymerase, an inhibitory effect of viral proteins on heterotypic virus function, or an inability to specifically incorporate heterotypic segments into budding virions. Here, we demonstrate that the full-length hemagglutinin (HA) of prototype influenza B viruses can complement the function of multiple influenza A viruses. We show that viral noncoding regions were sufficient to drive gene expression for either type A or B influenza virus with its cognate or heterotypic polymerase. The native influenza B virus HA segment could not be incorporated into influenza A virus virions. However, by adding the influenza A virus packaging signals to full-length influenza B virus glycoproteins, we rescued influenza A viruses that possessed HA, NA, or both HA and NA of influenza B virus. Furthermore, we show that, similar to single-cycle infectious influenza A virus, influenza B virus cannot incorporate heterotypic transgenes due to packaging signal incompatibilities. Altogether, these results demonstrate that the lack of influenza A and B virus reassortants can be attributed at least in part to incompatibilities in the virus-specific packaging signals required for effective segment incorporation into nascent virions.
IMPORTANCE Reassortment of influenza A or B viruses provides an evolutionary strategy leading to unique genotypes, which can spawn influenza A viruses with pandemic potential. However, the mechanism preventing intertypic reassortment or gene exchange between influenza A and B viruses is not well understood. Nucleotides comprising the coding termini of each influenza A virus gene segment are required for specific segment incorporation during budding. Whether influenza B virus shares a similar selective packaging strategy or if packaging signals prevent intertypic reassortment remains unknown. Here, we provide evidence suggesting a similar mechanism of influenza B virus genome packaging. Furthermore, by appending influenza A virus packaging signals onto influenza B virus segments, we rescued recombinant influenza A/B viruses that could reassort in vitro with another influenza A virus. These findings suggest that the divergent evolution of packaging signals aids with the speciation of influenza A and B viruses and is in part responsible for the lack of intertypic viral reassortment.

INTRODUCTION

Influenza A virus (IAV) and influenza B virus (IBV) are members of the Orthomyxoviridae family and have segmented genomes consisting of eight single-stranded, negative-sense viral RNA (vRNA) molecules (1). Influenza A viruses have a broad species tropism and mainly exist in the wild aquatic fowl reservoir, whereas influenza B viruses are primarily limited to the human population, although rare infections of seals have been documented (24). Despite these host reservoir differences, both influenza A and B viruses can cause severe infection in the human upper respiratory tract, leading to possible hospitalization or death, and are considered a major public health concern (1).
Influenza A and B viruses have similar genomes that encode homologous proteins but can be distinguished by the different lengths of proteins and noncoding regions that serve as promoters for replication and transcription (1). They are also distinguished by accessory proteins encoded from overlapping open reading frames (ORFs) and by the antigenic differences of internal proteins (5). For instance, influenza A and B viruses both encode ion channel proteins, M2 and BM2, respectively, whereas influenza A virus expresses the PB1–F2 pathogenicity factor and influenza B virus expresses the NB ion channel, which are absent in the converse virus (1). However, both influenza viruses encode two surface glycoproteins: hemagglutinin (HA), which is responsible for viral binding and fusion, and neuraminidase (NA), which is necessary for virus release from infected cells.
The HA and NA glycoproteins are the major antigenic determinants of influenza virus and are under immunologic pressure, leading to antigenic variants that are positively selected to avoid immune detection (6). A drastic change in antigenicity occurs during antigenic shift, which is caused by viral genome reassortment, or the transfer of genomic segments between different viral strains in coinfected cells within an organism (1). The antigenic diversity of the influenza virus glycoproteins is used to further classify influenza viruses, in which influenza A virus has 18 HA subtypes and 11 NA subtypes (1, 7, 8), and influenza B virus has two lineages (the Victoria-like and Yamagata-like lineages) that are divergent from the ancestral influenza virus B/Lee/1940 (B/Lee) strain and have been cocirculating in the human population since the 1980s (911). Influenza A and B viruses reassort intratypically between subtypes or lineages, but intertypic reassortment, or genetic swapping of segments between influenza A and B viruses, has never been observed (1, 9, 12, 13).
Considering the similarities between influenza A and B viruses and their cocirculation in the human population, the lack of intertypic reassortment is somewhat surprising. The mechanism governing this segregation is unclear, but intertypic incompatibility at the protein or vRNA level has been hypothesized to be an underlying factor (1416). Although recombinant chimeric influenza A/B viruses have been created, suggesting that portions of influenza B virus glycoproteins can replace the function of influenza A virus glycoproteins, substantial genetic modifications of the polypeptides were required for their successful rescue (1719). Additionally, the RNA-dependent RNA polymerase (RdRp) of influenza A or B virus has been shown to transcribe genes flanked by the heterotypic promoter sequences (2022) found in the terminal noncoding regions (NCRs) of each vRNA (23, 24). Influenza A virus vRNA can direct the incorporation of specific segments into budding virions, so that each nascent infectious virus contains a set of eight unique vRNA molecules (reviewed in reference 25). These sequence elements, referred to as packaging signals, are within the NCRs and coding regions of each influenza A viral segment and have not yet been characterized for influenza B viruses. Thus far, the mechanism governing how these sequence elements direct controlled packaging in influenza A virus and whether influenza B virus genomic packaging is directed by a similar mechanism remain unknown.
In order to gain further insight into the heterotypic incompatibilities between influenza A and B viruses, we used a single-cycle infectious influenza A virus (sciIAV) (26) to evaluate the ability of prototypic full-length influenza B virus HAs to replace the function of the influenza A virus counterpart. Using sciIAV, we observed efficient complementation with full-length influenza B virus HA protein. Our results were corroborated when we were able to generate a single-cycle infectious influenza B virus (sciIBV) where the single-cycle backbone dictates the transgene incorporation, likely via packaging signal recognition. Lastly, we rescued three viruses consisting of an influenza A virus backbone with either the HA, NA, or HA and NA of influenza B virus. To generate these viruses, the packaging signals for the respective influenza A virus glycoproteins were appended to the ORF of influenza B virus HA or NA (27, 28). This approach led to not only the first viable chimeric influenza A/B virus containing a full-length influenza B virus HA but also a chimeric influenza A/B virus containing full-length influenza B virus HA and NA, which retained the ability to reassort with a current circulating strain of influenza A virus.
Altogether, these results show that HA and NA of influenza A virus can be efficiently replaced by the corresponding influenza B virus glycoproteins and demonstrate that packaging signal compatibility plays an important role in the lack of intertypic reassortment, which suggests that speciation either led to or was caused by divergent packaging signal evolution.

MATERIALS AND METHODS

Cells and influenza viruses.

Human embryonic kidney 293T (293T) cells (ATCC CRL-11268) and Madin-Darby canine kidney (MDCK) cells (ATCC CCL-34) were maintained in Dulbecco's modified Eagle's medium (DMEM; Mediatech, Inc.) supplemented with 10% fetal bovine serum (FBS; Atlanta Biologicals) and 1% PSG (penicillin, 100 units/ml; streptomycin, 100 μg/ml; l-glutamine, 2 mM; Mediatech, Inc.). Cells were grown at 37°C in a 5% CO2 atmosphere. MDCK cells constitutively expressing HA (MDCK-HA) from the pandemic H1N1 virus A/California/4_NYICE_E3/2009 (pH1N1/E3) were previously described (4). MDCK-HA cells stably expressing HA from influenza virus A/Puerto Rico/8/1934 (H1N1; PR8), B/Lee, influenza virus B/Victoria/1/1987 (B/Vic), and influenza virus B/Yamagata/16/1988 (B/Yam) were generated by cotransfecting each pCAGGS HA plasmid and pCB7 (3:1 ratio) for eukaryotic expression of HA and hygromycin B resistance, respectively (4, 26, 29). HA-expressing cells were grown in DMEM–10% FBS–1% PSG supplemented with 200 μg/ml hygromycin B (Corning). After viral infections, cells were maintained at 33°C in a 5% CO2 atmosphere in DMEM containing 0.3% bovine serum albumin (BSA), 1% PSG, and 1 μg/ml tosylsulfonyl phenylalanyl chloromethyl ketone (TPCK)-treated trypsin (Sigma) (30).
Recombinant wild-type (rWT) PR8 (31), B/Yam (32), and pH1N1 (33) and chimeric influenza A/B virus in the genetic background of PR8 were rescued using ambisense reverse genetics plasmids (pDZ) as previously described (30, 34, 35). Influenza A virus X31 is a recombinant virus with a PR8 backbone that contains HA and NA from influenza virus A/Hong Kong/1/1968 (H3N2) (36). The sciIAV on the pH1N1 backbone expressing the green fluorescent protein (GFP) in place of HA has been previously described (4). Using the same approach, a sciIAV on the PR8 backbone was generated (31). To rescue sciIBV, influenza virus B/Brisbane/60/2008 (B/Bris) ambisense plasmids (pDP2002) were used as previously described (37). Virus stocks were propagated in MDCK (rWT) or MDCK-HA (sciIAV or sciIBV) cells for 3 days at 33°C in a 5% CO2 atmosphere (30). The virus titers of the stocks were determined by standard plaque assay (PFU/ml) in MDCK or MDCK-HA cells (4, 38). The virus titers in the tissue culture supernatants (TCSs) of multicycle growth analyses were determined by a focus formation assay (focus-forming units [FFU]/ml) by direct GFP quantification with fluorescence (sciIAV or sciIBV) or immunofluorescence (rWT) assays (4). Monoclonal antibodies (MAbs) against NP from influenza A virus (MAb HT103) (39) or influenza B virus (MAb B017; Abcam) and fluorescein isothiocyanate (FITC)-conjugated secondary antibodies were used, as described below and previously (4, 40).

Plasmids.

The reverse genetics plasmids used to generate PR8-sciIAV (31) and the influenza A virus ΔHA/GFP plasmid pPolI HA(45)GFP(80) have been previously described (28). The reverse genetics plasmids used to rescue B/Brisbane/60/2008 were generated by extracting RNA (kindly provided by the CDC) using an RNeasy minikit (Qiagen). Segments 1 to 3 and 7 were then cloned using a two-step reverse transcription-PCR (RT-PCR) strategy modeled after that used for influenza A virus (41, 42). Avian myeloblastosis virus reverse transcriptase and RNasin were used for the extension of vRNA as described by the manufacturer (Promega). For these segments, AarI and BsmB1 restriction sites were used to clone each segment into pDP2002 (43). Segments 4 to 6 and 8 were cloned according to the one-step RT-PCR strategy previously described (44). For all 8 segments, QuikLigase (New England BioLabs [NEB]) was used for ligation of the digested PCR product into pDP2002. Plasmid clones were sequenced by the Sanger sequencing method according to the BigDye Terminator kit protocol (BigDye Terminator [v3.1] cycle sequencing kit; Life Technologies) and run on a 3500xL genetic analyzer (ABI).
pCAGGS plasmids carrying B/Vic, B/Yam, and B/Lee HAs were generated by RT-PCR from RNA isolated from infected MDCK cells, using the EcoRI and NheI restriction enzymes (NEB). Minigenome (MG) plasmids containing GFP or firefly luciferase (FFluc) flanked by the 3′ and 5′ NCRs of the influenza B virus HA segment (IBV MG) were cloned into the human pPolI plasmid using SapI (NEB), as previously described for the IAV MG plasmid (31, 45). Ambisense pDZ plasmids carrying PR8 or B/Yam PB2, PB1, PA, and NP or pCAGGS carrying lymphocytic choriomeningitis virus (LCMV) NP were previously described (31, 32, 46). Chimeric influenza A/B virus constructs were generated by subcloning the entire ORF of B/Yam HA or NA into vRNA expression plasmids that contained the 3′ and 5′ NCRs and packaging signals from influenza A virus HA (28) or NA (47). B/Yam HA was cloned into human pPolI HA(45)GFP(80) (28) using NheI and XhoI (NEB), and B/Yam NA was cloned into pHW2000 Flu-GFP (47) using MluI and SpeI (NEB). Influenza B virus ΔHA/GFP plasmid pPolI HA(228)GFP(108) was generated as previously described (28). The B/Yam HA segment was amplified by RT-PCR from RNA isolated from infected MDCK cells and cloned into the human pPolI plasmid using SapI (NEB) and then digested with BglII and BsaI (NEB) to cut 228 nucleotides (nt) from the 3′ end and 108 nt from the 5′ end. The product from PCR amplification of GFP using primers flanked by BglII and BsaI was subcloned between these two restriction sites. Following rescue of sciIBV (228)GFP(108), sequencing revealed that pPol HA(228)GFP(108) contained a primer duplication mutation. This mutation introduced the primer twice (on the 3′ end) immediately upstream of the GFP ORF. All plasmids were generated using standard cloning techniques and purified using a Wizard SV kit (Promega). The primers (Integrated DNA Technologies, Inc.) used for the generation of the described plasmid constructs are available upon request. All plasmid constructs were verified by DNA sequencing (ACGT, Inc.).

Immunofluorescence assay (IFA).

For the characterization of MDCK-HA cells, confluent monolayers of parental MDCK or MDCK-HA cells (105 cells, 48-well plate format) were fixed with 4% paraformaldehyde for 15 min at room temperature and blocked overnight with 2.5% BSA at 4°C. Cells were then incubated with mouse MAb 31C2 or 29E3 (pH1N1 HA, 1 μg/ml) (48), MAb PY102 (PR8 HA, 1.5 μg/ml) (49), or a 1:1,000 dilution of goat polyclonal antibody (PAb) against HA of influenza virus B/Hong Kong/8/1973 (PAb NR-3165; NIAID Biodefense and Emerging Infectious Research Resources Repository [BEI Resources]) for 1 h at 37°C. After washing with 1× phosphate-buffered saline (PBS), the cells were incubated with FITC-conjugated secondary antimouse (1:140; Dako) or antigoat (1:200; Jackson ImmunoResearch) antibodies and 4′,6′-diamidino-2-phenylindole (DAPI; Research Organics) for 30 min at 37°C. After three washes with 1× PBS, the cells were visualized using a fluorescence microscope, photographed (Cooke Sensicam QE) with a ×20 objective (actual magnification, ×200), and colored and merged using Adobe Photoshop CS4 (v11.0) software. Representative images from at least three independent fields are shown.
For virus characterization, confluent monolayers of MDCK cells (105 cells, 48-well plate format) infected at a multiplicity of infection (MOI) of 10 were fixed at 8 h postinfection as described above and permeabilized with 0.2% Triton X-100. Viral HA or NP was detected using antibodies PY102 and HT103, respectively, for influenza A virus or antibodies NR-3165 and B017, respectively, for influenza B virus. NA was stained with mouse anti-PR8 MAb NR-4540 (1:500 dilution; BEI Resources) for influenza A virus and goat anti-B/Lee PAb NR-3114 (1:1,000 dilution; BEI Resources) for influenza B virus and stained with FITC-conjugated secondary antibodies and DAPI, as described above. For phenotypic analysis of influenza A/B reassortant viruses, 1 μg/ml of mouse MAbs against influenza A virus HA (29E3), NA (10C9), nonstructural region 1 (NS1; 1A7), or NP (NR-4545) or goat PAbs against influenza B virus HA (NR-3165) or NA (NR-3114) were used as described above.

Flow cytometry.

Parental MDCK and MDCK-HA cells (106 cells, 6-well plate format) were trypsinized and stained in suspension as previously described (4). Briefly, cells were washed and incubated with 31C2, PY102, NR-3165, rabbit anti-B/Lee PAb (V-63, 1:1,000) (18), and mouse anti-B/Yam MAb (MAb 15B6, 1:1 of hybridoma TCS) (18) for 30 min at 4°C. After washing 3 times in staining buffer (1× PBS with 5% FBS). cells were stained with FITC-conjugated secondary antimouse or antirabbit antibodies (1:140; Dako) for 30 min at 4°C. Cells were washed 3 times with staining buffer, and the percentage of fluorescent cells was determined using a C6 4-color flow cytometer (Accuri). The frequencies of FL1-positive cells that fell into an appropriate side scatter gate were analyzed using FlowJo software (Tree Star). Parental cells stained with antibodies were used as a negative control. To evaluate MG activity, 293T cells were transfected as described below and 48 h later were trypsinized, washed 3 times in staining buffer, and analyzed using a C6 flow cytometer. The percentages and mean fluorescence intensities (MFIs) of GFP-positive (GFP+) cells were determined by gating on FL1-positive cells.

Protein gel electrophoresis and Western blot analysis.

Parental MDCK or MDCK-HA cells were evaluated for protein expression by Western blotting as previously described (50). Briefly, samples were lysed in passive lysis buffer (Promega), separated on a 10% SDS-polyacrylamide gel, and transferred onto nitrocellulose membranes (Bio-Rad) for 2 h at 175 mA. After blocking for 1 h at room temperature with 10% dry milk in 1× PBS–0.1% Tween 20, membranes were incubated with antibodies against HA (31C2, 1 μg/ml; PY102, 1 μg/ml; NR-3165, 1:1,000) or mouse monoclonal antiactin antibody (1:1,000; Sigma) as a loading control. The membranes were then washed 3 times with 1× PBS–0.1% Tween 20 and probed with secondary horseradish peroxidase (HRP)-conjugated anti-mouse or anti-goat Ig (1:2,000 dilution; GE Healthcare) for 1 h at room temperature. After washing 3 times with 1× PBS–0.1% Tween 20, protein bands were detected using a chemiluminescence kit (Denville Scientific Inc.) and photographed using a Kodak ImageStation.

Viral growth kinetics.

Multicycle growth analyses were performed by infecting confluent monolayers of parental MDCK or MDCK-HA cells (5 × 105 cells, 12-well plate format) at an MOI of 0.001 (in triplicate), and the cells were incubated at 33°C for 3 days (26). At the times postinfection indicated below, GFP expression was assessed by fluorescence microscopy, and viral titers in the TCS were measured by evaluating the number of FFU/ml in a focus formation assay. Briefly, confluent wells of MDCK-HA cells (5 × 104 cells, 96-well plate format, in triplicate) were infected with 10-fold serial dilutions of the TCS. At 18 h postinfection, the cells were washed with 1× PBS and foci were visualized using a fluorescence microscope and enumerated. Titrations of chimeric influenza A/B viruses were performed in a manner similar to that described above, but individual infected cells were detected at 10 h postinfection by immunofluorescence using influenza virus NP MAb HT103 (PR8) or B017 (B/Yam). Mean values and standard deviations (SDs) were calculated using Microsoft Excel software. The limit of detection was determined by the initial dilution of TCS for titration, where 50 μl of a 1:10 dilution was used.

Plaque assays.

The plaque phenotype of PR8 or pH1N1 sciIAVs in parental MDCK or MDCK-HA cells was determined by infecting confluent monolayers of cells (106 cells, 6-well plate format) with specified viruses for 3 days at 33°C in postinfection medium containing agar. Monolayers were then fixed with 4% paraformaldehyde for 15 min at room temperature and stained with 0.1% crystal violet (Fisher Scientific). Plaque assays for chimeric influenza A/B viruses were performed similarly, but after infection and fixation, the monolayers were permeabilized and immunostained as previously described (29) using HT103 or B017 to detect influenza A or B virus NP, respectively.

HAI assays.

Receptor-destroying enzyme-treated (Denka Seiken) sera or MAbs were serially diluted in 96-well V-bottom plates and mixed 1:1 with 8 hemagglutinating units (HAU) of influenza A or B virus HA-pseudotyped PR8-sciIAV or rWT PR8, X31, or B/Yam for 30 min at room temperature. Hemagglutination inhibition (HAI) assay titers were determined by adding 0.5% turkey red blood cells (RBCs) to the virus-antibody mixtures for 30 min on ice, as previously described (51). For sciIAV HAI assays, MAbs 31C2, PY102, NR-3165, and V-63 or PAbs anti-B/Vic and anti-B/Yam sheep (which were obtained from the CDC and which were for the years 2012 and 2013) were used.

MG assays.

Human 293T cells were transfected in suspension (5 × 105 cells, 12-well plate format, in triplicate) with 500 ng each of influenza A or B virus ambisense pDZ PB2, PB1, and PA (3P) plasmids in the absence (negative control) or presence of the respective pDZ NP plasmid, together with influenza A or B virus MG plasmids expressing GFP or FFluc. A plasmid expressing Renilla luciferase under the control of the simian virus 40 (SV40) promoter (Promega) was cotransfected to normalize transfection efficiencies (52). In all cases, cells were cotransfected with empty pDZ plasmid to achieve equimolar DNA concentrations. Lipofectamine 2000 transfection reagent (Invitrogen) was used at a 1:1 ratio with plasmid DNA. At 48 h posttransfection, cells were prepared for flow cytometry or cell lysates were prepared using passive lysis buffer (Promega). Renilla luciferase and FFluc expression was determined using Stop & Glow reagents (Promega) and quantified with a Lumicount luminometer (Packard). The fold induction over the level of induction for the negative controls was calculated, and statistics (Student's two-tailed t test) were calculated with GraphPad Prism software. For competition experiments, increasing concentrations (250, 500, or 1,000 ng) of competing PR8, B/Yam, or LCMV NP expression plasmids were added, in addition to 3P plus NP and MG vRNA expression plasmids, and values were normalized to the fold induction in the absence of competitors (percent reporter expression).

Evaluation of vRNA incorporation into virions.

Rescue transfections where sciIAV or sciIBV was each rescued with the ΔHA/GFP construct pPolI influenza A virus HA(45)GFP(80) or pPolI influenza B virus HA(228)GFP(108) were performed as previously described (26). Briefly, 293T cells (106 cells, 6-well plate format) were cotransfected with seven ambisense rescue plasmids (for PB2, PB1, PA, NP, NA, M, and NS) for PR8 (31) or B/Bris (37), together with pCAGGS HA protein expression plasmids to facilitate initial rescue (26), and ΔHA/GFP vRNA expression plasmids, using Lipofectamine 2000 (1:1 ratio). The GFP fluorescence from vRNA expression plasmids in rescue transfections was monitored at 2 days posttransfection. Directly afterward, TCSs were collected and passaged onto fresh HA-expressing MDCK cells. The rescue of sciIAV or sciIBV with each ΔHA/GFP construct was confirmed by fluorescence and standard hemagglutination assay at 2 to 4 days postinfection (30). Triplicate transfections were performed in five independent experiments.

Mouse experiments.

For immunogenicity and pathogenicity experiments, female 6- to 8-week-old C57BL/6 mice were purchased from the National Cancer Institute (NCI) and maintained at the University of Rochester under specific-pathogen-free conditions. All mouse experiments were approved by the University Committee of Animal Resources and complied with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Research Council (53). Infections were performed as previously described (4). Briefly, mice were anesthetized intraperitoneally (i.p.) with 2,2,2-tribromoethanol (Avertin; 240 mg/kg of body weight) and inoculated intranasally (i.n.) with 30 μl of virus preparations (4). Morbidity was monitored by determination of the percentage of body weight loss relative to the starting weight. Mice losing greater than 25% of their body weight were euthanized humanely. At 2 weeks postinfection, mouse sera were collected by submandibular bleeding and stored at −20°C. Naive serum was collected from animals prior to infection and used as a negative control. The 50% mouse lethal dose (MLD50) was determined by infecting five groups of mice (n = 4) with 10-fold serial dilutions of 105 to 101 PFU and calculated using the method of Reed and Muench (54).

ELISA.

Enzyme-linked immunosorbent assays (ELISAs) were performed as previously described by coating plates for 18 h at 4°C with lysates from PR8- or B/Yam-infected MDCK cells (4). After blocking with 1% BSA, the plates were incubated with 2-fold dilutions of serum for 1 h at 37°C (1:250 starting dilution), washed with H2O, and incubated with HRP-conjugated goat anti-mouse IgG (1:2,000; Southern Biotech) for 30 min at 37°C. Reactions were then developed with tetramethylbenzidine substrate (BioLegend) for 5 min at room temperature, quenched with 2 N H2SO4, and read at 450 nm (Vmax kinetic microplate reader; Molecular Devices).

RESULTS

Full-length influenza B virus HA complements HA-deficient sciIAV.

The minimal requirement for intertypic virus reassortment is conservation of protein function. To determine the functional compatibility of influenza B virus HA with an influenza A virus backbone, we chose to utilize our HA-deficient sciIAV approach (26). This virus requires HA protein trans-complementation, because the fourth segment is modified to encode GFP (28). MDCK cells that stably express full-length influenza virus type A (pH1N1/E3 and PR8) or type B (B/Lee, B/Vic, and B/Yam) HA proteins were first generated (Fig. 1). All MDCK-HA cells expressed the viral glycoproteins on the cell surface (Fig. 1A) at comparable levels (Fig. 1B). Flow cytometric detection of HA then demonstrated the distinct antigenic signature of the various MDCK-HA stable cell lines (Fig. 1C).
FIG 1
FIG 1 Characterization of influenza B virus HA-expressing MDCK cells. (A) HA protein detection by indirect immunofluorescence. HA-expressing MDCK cells were fixed and stained with influenza A virus HA MAbs 31C2 (pH1N1) and PY102 (PR8) or anti-influenza B virus HA PAb NR-3165 and counterstained with DAPI to visualize the nuclei. Parental MDCK cells were used as a negative control. Representative images obtained with a ×20 objective (actual magnification, ×200) are shown. Bars, 20 μm. (B) Western blotting results. Parental and HA-expressing MDCK whole-cell lysates were stained with MAbs 31C2 and PY102 and the PAb NR-3165. MAb against actin was used as a loading control. (C) Flow cytometry. Parental and HA-expressing MDCK stable cell lines infected with pH1N1/E3, PR8, B/Lee, B/Vic, and B/Yam were detected with antibodies 31C2, PY102, and NR-3165, an MAb generated against B/Yam (15B6), or an anti-B/Lee PAb (V-63). The percentage of positively stained cells is indicated (y axis).
To evaluate intertypic HA compatibility, sciIAV was used to infect MDCK-HA cells, where productive infection can result only from functional HA complementation. An sciIAV that contained seven segments of PR8 plus the influenza A virus HA(45)GFP(80) transgene was used to infect MDCK-HA cells at a low MOI, and any virus progeny would be pseudotyped with HA from the cells (Fig. 2) (28). Regardless of homo- or heterotypic HA expression, the MDCK-HA cells were able to complement the HA-deficient, GFP-expressing sciIAV over multiple cycles of infection, as characterized by fluorescence microscopy that showed the spread of GFP expression (Fig. 2A) and the production of infectious progeny in the TCS (Fig. 2B). As expected, no propagation of virus was seen in parental MDCK cells (26). These results were corroborated by plaque assays (Fig. 2C). Similar trans-complementation was achieved when cells were infected with an evolutionarily distinct sciIAV containing the backbone of the pH1N1 reassortant virus that emerged in 2009 (4), which possesses internal segments of both avian and swine influenza virus origin (Fig. 3) (55). Influenza virus type A or B MDCK-HA cells also complemented the growth of pH1N1-sciIAV, as shown by the spread of GFP expression (Fig. 3A), virus production (Fig. 3B), and the plaque size phenotype (Fig. 3C). The identity of the MDCK-HA cells was further confirmed by performing HAI assays of the HA-pseudotyped sciIAV using MAbs and PAbs specific for each virus isolate (Table 1). Together, these results indicate that full-length influenza B virus HA protein complements HA-deficient sciIAV to generate an infectious influenza virus.
FIG 2
FIG 2 Mouse-adapted PR8-sciIAV is complemented by HA proteins from prototypic influenza B virus lineages. (A and B) Multicycle growth analysis of PR8-sciIAV in parental MDCK and MDCK-HA cells. Confluent monolayers of parental and HA-expressing MDCK cells were infected (in triplicate) with PR8-sciIAV at a low MOI (0.001). (A) At various times postinfection, GFP was visualized by fluorescence microscopy (with a ×10 objective; magnification of actual representative images, ×100). Bars, 40 μm. (B) TCSs were collected for titration in MDCK-HA cells. Data represent the means ± SDs of the results determined for triplicate assays. Dashed black line, limit of detection (200 FFU/ml). (C) Plaque morphology of PR8-sciIAV in parental MDCK and MDCK-HA cells. Parental and HA-expressing MDCK cell lines were infected with 50 PFU of PR8-sciIAV, and at 3 days postinfection, monolayers were stained with crystal violet.
FIG 3
FIG 3 Pandemic 2009 pH1N1-sciIAV is complemented by influenza B virus HA-expressing cell lines. (A and B) Multicycle growth analysis of pH1N1-sciIAV in parental MDCK and MDCK-HA cells. Triplicate confluent monolayers of parental and HA-expressing MDCK cells were infected with pH1N1-sciIAV (MOI, 0.001). (A) At various times postinfection, GFP was visualized by fluorescence microscopy (with a ×10 objective; magnification of actual representative images, ×100). Bars, 40 μm. (B) TCSs were collected for titration in MDCK-HA cells. Data represent the means ± SDs of the results determined for triplicate assays. Dashed black line, limit of detection (200 FFU/ml). (C) Plaque morphology of pH1N1-sciIAV. Parental and HA-expressing MDCK cell lines were infected with 50 PFU of pH1N1-sciIAV, and at 3 days postinfection, monolayers were stained with crystal violet.
TABLE 1
TABLE 1 HAI activity of specific MAbs and PAbs against IAV or IBV HA-pseudotyped sciIAV
Antibody HAI neutralization titera
pH1N1/E3 PR8 B/Lee B/Vic B/Yam
31C2 54.7        
PY102   58.6      
NR-3165 <20 <20 320 640 160
V-63 <20 <20 640 40 40
Anti-B/Vic <20 <20 40 160 <20
Anti-B/Yam <20 <20 40 <20 160
a
Eight HAU of the indicated viruses was incubated in triplicate with 2-fold serial dilutions of the indicated antibodies. MAbs were 31C2 and PY102 (for which the titers are in ng/ml), and PAbs were NR-3165, V-63, anti-B/Vic, and anti-B/Yam (for which titers are reciprocal dilutions).

Influenza A and B viruses share promoter and polymerase specificity.

In a previous study, Muster and colleagues demonstrated that a replicating influenza A virus could be rescued when the NCR of influenza A virus NA was replaced with the NCR of the influenza B virus nonstructural (NS) segment (20). This demonstrated that influenza A virus RdRp can utilize the promoter activity of influenza B virus NCR, but importantly, the regions later to be defined as packaging signals (within the ORF) were left unchanged. In order to more fully define the NCR and RdRp compatibilities between influenza A and B viruses (Fig. 4), we used type A virus MG plasmids (52) to generate similar virus-like RNA expression plasmids that transcribe GFP or FFluc, which is flanked by influenza B virus NCRs (Fig. 4A). To normalize for cellular translation levels, a protein expression plasmid encoding Renilla luciferase under the control of the SV40 promoter was cotransfected with the MG plasmids. By flow cytometry, a significantly higher percentage of cells was found to express GFP from the MG when homotypic RdRp rather than heterotypic RdRp was cotransfected (Fig. 4B). Additionally, homotypic MG activity was greater in GFP+ cells, as the MFI was higher (Fig. 4B). Luminescence quantification of MG activity provided similar results, where influenza A or B virus RdRp could transcribe the heterotypic MG, albeit it transcribed the heterotypic MG to lower levels than it transcribed the homotypic MG (Fig. 4C). Although heterotypic RdRp is not as efficient as homotypic RdRp for MG replication and transcription, these data suggest that the lack of type A and B virus heterotypic reassortment is not due to an incompatibility of promoters or polymerase activities.
FIG 4
FIG 4 Promoter and polymerase compatibility of type A and B influenza viruses. (A) Schematic representation of influenza A and B virus MG constructs. GFP or FFluc MG plasmids driven by the influenza A virus (white box) or influenza B virus (black) NCRs are indicated. (B and C) MG assays. Human 293T cells were cotransfected with 500 ng of ambisense pDZ expression plasmids encoding the minimal requirements for viral replication and transcription (3P and NP), together with MG vRNA-like expression plasmids encoding GFP (B) or FFluc (C) flanked by influenza A or B virus NCRs (IAV MG and IBV MG, respectively) plus polymerase II-driven Renilla luciferase (C) plasmids as a transfection control. At 2 days posttransfection, cells were prepared for flow cytometry to determine the percentage and MFI of GFP+ cells (B) or lysed for luminescence evaluation (C). FFluc activity was normalized to that of Renilla luciferase. Data represent the means ± SDs of the results determined for triplicate assays. (D) Influenza B virus NP inhibits IAV MG activity. An IAV or IBV MG with the corresponding 3P plus NP was transfected as described above in the absence of NP (−) or in the presence of 250, 500, or 1,000 ng of competing protein expression plasmids encoding heterotypic influenza virus NP or LCMV NP. Normalized reporter expression provided (in percent) is relative to MG activity in the absence of competing NP. Statistical analysis was performed using Student's two-tailed t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Other discrepancies that might impede heterotypic reassortment could include the inhibitory effect of influenza B virus NP on influenza A virus replication (56). Indeed, we observed that coexpression of influenza B virus NP inhibited IAV MG expression, whereas that of the NP of another negative-stranded RNA virus (LCMV) did not (Fig. 4D). Whether this inhibition results from affinity differences and substrate (vRNA/cRNA) binding competition, a specific virus, host protein interaction with influenza B virus NP, or some other mechanism is unknown. However, even in systems devoid of influenza B virus NP, virus rescue still does not occur when the HA segment of influenza B virus is used to complement reverse genetics plasmids encoding seven segments of influenza A virus (18, 19), suggesting that a separate mechanism is responsible for the lack of intertypic reassortants in the absence of this protein.

Specific incorporation of transgenes by sciIAV and sciIBV requires homotypic packaging signals.

The packaging signals of influenza A virus have been studied extensively (27, 28, 5767), yet such nucleotide signals have not been described for influenza B virus. Using an approach that added various lengths of the 3′- and 5′-terminal coding region of influenza B virus HA to a GFP gene, we generated an IBV HA(228)GFP(108) vRNA construct that was routinely incorporated into influenza B virus virions in the presence of exogenously expressed HA protein (Fig. 5A). Using reverse genetics plasmids and influenza B virus-infected MDCK-HA cell lines, we rescued a sciIBV. To assess the ability of influenza A or B virus to incorporate a heterotypic transgene, we tested if sciIAV could incorporate the influenza B virus HA(228)GFP(108) vRNA or if sciIBV could incorporate the influenza A virus HA(45)GFP(80) vRNA (Fig. 5B). While the RdRp of the influenza A or B viruses could replicate and transcribe GFP from either type A or type B influenza virus vRNAs (Fig. 5C), sciIAV could incorporate only the influenza A virus HA(45)GFP(80) construct and sciIBV could incorporate only the influenza B HA(228)GFP(108) construct when rescue transfection TCSs were passaged onto fresh MDCK-HA cells (Fig. 5D). This observation was verified by five independent experiments, each including triplicate transfections (Fig. 5E). These results suggest that packaging signals play an important role in the specific incorporation of type A or B influenza virus vRNAs into nascent virions.
FIG 5
FIG 5 Type-specific packaging signals dictate segment incorporation into influenza A or B virions. (A) Schematic representation of IAV and IBV vRNA segment 4 constructs. Lines, NCRs; boxes with lines, packaging signals flanking the GFP ORF. (B) Rescue transfection and infection diagram. Ambisense pDZ expression plasmids containing seven different segments of influenza A or B virus were cotransfected together with the GFP vRNA pPolI expression plasmid (A) and the HA protein expression plasmid pCAGGS into 293T cells. At 48 h posttransfection, TCSs were collected and used to infect fresh monolayers of MDCK HA-expressing cells to evaluate virus rescue. (C) Fluorescence microscopy of rescue transfections. GFP expression from transfected human 293T cells was evaluated under a fluorescence microscope at 72 h posttransfection. (D) Virus rescues. TCSs from rescue transfections (C) were clarified and used to infect fresh MDCK-HA cells. At 48 h postinfection, infected cells were visualized for the presence of GFP under a fluorescence microscope. Representative images obtained with a ×10 objective are shown (actual magnification shown, ×100). Bars, 40 μm. (E) Virus rescues. Attempts to rescue intertypic sciIV were performed five times in triplicate. The percent rescue efficiency is indicated.

Full-length influenza B virus glycoproteins that contain influenza A virus packaging signals are incorporated into influenza A virus virions.

To query if cis-acting packaging signals from influenza A virus were sufficient for incorporation of influenza B vRNAs into influenza A viruses, we performed gain-of-function experiments (Fig. 6). Using a strategy similar to that used to generate sciIAV (26, 47), we constructed chimeric influenza A/B virus genomic segments, where the entire ORF of B/Yam HA or NA replaced that of influenza A virus, except that the type A packaging signals and NCRs were left unchanged (Fig. 6A). Plasmids encoding the chimeric influenza A/B virus segments with the remaining PR8 reverse genetics plasmids were used to rescue recombinant virus containing HA, NA, or HA plus NA of influenza B virus (here referred to as PR8 BHA, BNA, and BHANA, respectively). The identities of the replication-competent, plaque-purified recombinant viruses were confirmed by RT-PCR (data not shown) and immunofluorescence of infected cells (Fig. 6B).
FIG 6
FIG 6 Manipulation of influenza A virus packaging signals to generate chimeric glycoprotein influenza A/B viruses. (A) Schematic representation of viral segments encoding HA and NA. Influenza type A virus vRNA (white boxes) with packaging signals (boxes with lines) and influenza type B virus vRNA (black boxes) are indicated. The BHA or BNA chimeric vRNA constructs include the influenza A virus NCR (lines) and packaging signals (boxes with lines), together with the complete ORFs of the influenza B virus HA and NA glycoproteins, respectively. The length of each vRNA segment (in nucleotides) is indicated on the right. (B) Antigenic characterization of recombinant PR8 viruses containing BHA, BNA, or both BHA and BNA (BHANA). MDCK cells were infected with the indicated viruses (MOI, 0.1) for 16 h, and IAV and IBV type-specific HA, NA, or NP antibodies were used to detect protein expression (green). Cell nuclei were stained with DAPI (blue). Anti-influenza A virus antibodies were PY102 for HA, NR-4540 for NA, and HT103 for NP. Anti-influenza B virus antibodies were NR-3165 for HA, NR-3114 for NA, and B017 for NP. Representative images obtained with a ×20 objective are shown (actual magnification shown, ×200). Bars, 20 μm. (C) Multicycle growth analysis of chimeric type A/B viruses. Confluent monolayers of MDCK cells (in triplicate) were infected with the indicated viruses (MOI, 0.001), and at the indicated times postinfection, TCSs were collected for titration in MDCK cells. Data represent the means ± SDs of the results determined for triplicate assays. Dashed black line, limit of detection (200 FFU/ml). (D) Plaque morphology of chimeric influenza A/B viruses in MDCK cells. Monolayers of MDCK cells were infected with the PR8 and B/Yam viruses and chimeric glycoprotein influenza A/B viruses (PR8 HA, PR8 NA, and PR8 HANA). At 3 days postinfection, monolayers were immunostained with influenza A virus (PR8, PR8 BHA, PR8 BNA, and PR8 BHANA) or influenza B virus (B/Yam) NP monoclonal antibodies HT103 and B017, respectively.
With the rescued type A viruses containing the glycoproteins of influenza B virus, we sought to characterize any effects on virus fitness. PR8 BHA and BNA have replication kinetics similar to those of rWT PR8, but PR8 BNA peak titers were moderately lower. In contrast, PR8 BHANA displayed slightly delayed kinetics comparable to those of rWT B/Yam, which grows slower than PR8 (Fig. 6C). Analysis of the plaque size phenotype provided results that agree with the observations presented above (Fig. 6D). Both the production of recombinant chimeric influenza virus and the efficient in vitro replication of PR8 BHA, BNA, and BHANA indicate that manipulation of influenza A virus packaging signals is sufficient to incorporate an influenza B viral segment with an homologous function and can allow the generation of intertypic reassortant viruses.

Pathogenicity and immunogenicity of influenza A/B viruses with recombinant glycoproteins.

To evaluate the ability of chimeric influenza A/B viruses with recombinant glycoproteins to thrive in vivo, we sought to first test the morbidity induced by intranasal infection of mice (Fig. 7). A dose of 105 PFU was selected for all viruses except PR8 (10 PFU), because of the mild pathogenicity of influenza B virus in C57BL/6 mice (68). The morbidity phenotype of PR8 BHANA mirrored that of B/Yam, as determined by body weight loss (<5%), and infection with PR8 BHANA did not lead to any mortality, whereas infection with PR8 BHA led to moderate weight loss (>15%). In contrast, PR8 BNA at the same dose was fatal to mice, with a calculated MLD50 of 50 PFU (Fig. 7B), which is only twice that of rWT PR8 (MLD50, ∼25 PFU) (68) and suggests that PR8 HA is a major pathogenicity factor in mice, similar to the results found for the pandemic 1918 Spanish influenza virus (69).
FIG 7
FIG 7 Pathogenicity and immunogenicity of influenza A/B viruses with recombinant glycoproteins. (A) Morbidity caused by influenza A/B viruses with chimeric glycoproteins. Female 6- to 8-week-old C57BL/6 mice were inoculated intranasally with 105 PFU of influenza B virus glycoprotein-containing viruses (B/Yam, PR8 BHA, PR8 BNA, or PR8 BHANA) or 10 PFU of PR8 (n = 4). For 2 weeks postinfection, weight loss was monitored daily. (B) Lethality of PR8 BNA. Tenfold decreasing doses or PR8 BNA were used to infect 6- to 8-week-old female C57BL/6 mice intranasally (n = 4). The MLD50 was determined using the method of Reed and Muench (54). (C and D) Humoral immune responses. Total anti-influenza virus IgG antibodies in serum collected at 2 weeks postinfection with the indicated viruses were determined by ELISA (n = 4). Plates were coated with lysates from PR8-infected (C) or B/Yam-infected (D) MDCK cells. Data represent the means ± SEMs of the results determined for individual mice. OD, optical density; Ag, antigen.
To demonstrate the immunogenicity of influenza A/B viruses with recombinant glycoproteins, systemic IgG antibodies were evaluated in mice infected with sublethal doses of virus. As expected, chimeric and rWT viruses induced comparable amounts of total anti-influenza A antibodies, as determined by ELISA (Fig. 7C). This outcome is perhaps due to saturation of the assay by non-HA and non-NA antibodies, such as anti-NP antibodies (70). When evaluating anti-influenza B virus antibodies, we found that only chimeric viruses carrying influenza B virus HA (PR8 BHA and PR8 BHANA) induced detectable levels of total anti-influenza B virus antibodies (Fig. 7D), suggesting that type B virus NA was not strongly immunogenic in this model. To validate our approach as a potential vaccine platform, we showed that antibodies from mice infected with influenza A/B viruses with recombinant glycoproteins had potent HAI activity (71) against homologous virus (Table 2). These strategies, coupled with intrinsic and further attenuating mutations, such as those found in the live attenuated influenza vaccine (72), could be synergized to generate a safe, immunogenic, multivalent vaccine against type A and B viruses using a single type A vaccine virus backbone.
TABLE 2
TABLE 2 HAI titers of mouse sera inoculated with chimeric influenza A/B viruses
Virus inoculumb Geometric mean (SD) serum HAI titera
PR8 X31 B/Yam
Naive <16 (0) <16 (0) <16 (0)
PR8 256 (161) <16 (0) <16 (0)
B/Yam88 <16 (0) <16 (0) 215 (64)
PR8 BHA <16 (0) <16 (0) 431 (128)
PR8 BNA 215 (64) <16 (0) <16 (0)
PR8 BHANA <16 (0) <16 (0) 215 (64)
a
Eight HAU of the indicated viruses was incubated with a 2-fold serial dilution of the indicated serum.
b
Anesthetized mice were administered virus intranasally (n = 4), and serum was collected at 14 days postinfection.

Reassortment of influenza B virus glycoproteins containing influenza A virus packaging signals.

Reassortment occurs readily within the different influenza virus types; thus, the ability of chimeric influenza A/B virus segments to move between homotypic genetic backbones during coinfection was analyzed (Fig. 8). To both assess reassortment ability and verify that chimeric influenza A/B glycoproteins can also function in pH1N1 virus, we coinfected MDCK cells with equivalent amounts of rWT pH1N1 and PR8 BHANA (MOI, 3). At 16 h postinfection, TCS was collected to isolate individual virus clones by plaque assay and to identify reassortment events using IFA against influenza B virus HA or NA or influenza A virus HA, NA, NP, or NS1 (Fig. 8A). Viruses that possessed either the influenza B virus HA or the influenza B virus NA in the context of novel constellations of PR8 or pH1N1 segments were identified (Fig. 8B). In agreement with the generation of PR8 BHA and PR8 BNA, we observed the compatibility of influenza B virus HA or NA with pH1N1, suggesting that the enzymatic and functional activities of these proteins were functional. For instance, one of the isolated viruses contained the viral HA from pH1N1, while NP, NA, and NS were derived from PR8 BHANA (phenotype 1). Another viral isolate contained similar segment constellations, but in this case the NS segment was derived from pH1N1, as determined by the lack of recognition with the PR8 NS1 MAb 1A7 (phenotype 2) (73). Finally, a virus containing HA, NP, and NS of PR8 HANA and NA from pH1N1 was also identified (phenotype 3). Altogether, these results suggest that the packaging signals of influenza A virus are sufficient in granting intertypic reassortment when the genes, including those from influenza B viruses, possess homologous functions and are at least in part responsible for the lack of reassortment between influenza A and B viruses.
FIG 8
FIG 8 Chimeric influenza A/B virus can reassort with influenza A virus. (A) Experimental design. MDCK cells were coinfected with pH1N1 and PR8 BHANA (MOI, 3) for 16 h at 33°C. Viruses from TCSs were plaque purified, amplified once on MDCK cells, and used to infect cells to determine the phenotype (ph.) by IFA. Relative segment sizes (in descending order, PB2, PB1, PA, HA, NP, NA, M, and NS) and colors (black, pH1N1; blue, PR8; red, B/Yam) represent identity. (B) Phenotypes of three selected reassortant viruses. MDCK cells were infected with parental or plaque-purified reassortant viruses (MOI, 0.1; 24 h; 33°C) and phenotypically characterized by IFA. The antibodies 29E3 and NR-3165 were used to detect IAV and IBV HAs, respectively; 10C9 and NR-3114 were used to detect IAV and IBV NAs, respectively; 1A7 was used to detect PR8-specific NS1; and NR-4545 was used to detect PR8-specific NP. Representative images obtained at ×20 magnification are shown (actual magnification, ×200). Bars, 20 μm.

DISCUSSION

The evolutionary divergence of type A and B influenza viruses over 4,000 years ago (74) has been followed by further adaptation of these viruses within their natural reservoir of wild aquatic fowl and humans, respectively (75). Through this diversification, however, both viruses have retained similar genomes, replication cycles, and general protein and enzymatic functions (1). Despite these similarities, no reassortments between influenza A and B viruses have been described to date. Several mechanisms may prevent intertypic reassortment: inefficient protein-protein interactions, an inability of RdRps to replicate and transcribe heterotypic viral segments, specific inhibition of influenza A virus by influenza B virus NP, or, as demonstrated in this report, incompatible packaging signals that impede heterotypic gene incorporation into virions.
We demonstrate here that despite evolutionary divergence, full-length influenza B virus HA expressed by cell lines (Fig. 1; Table 1) can complement HA-deficient PR8 (Fig. 2) and pH1N1 (Fig. 3) sciIAVs at the protein level. These results are in contrast to those of three previous studies where the signal peptide (19) or transmembrane and cytoplasmic tail domains (18, 19, 76) of influenza A virus HA were required for the rescue of high-titer influenza A viruses expressing influenza B virus HA. In the former case, the authors suspected that packaging signals played a role because after cleavage, the signal peptide is not thought to affect the virus life cycle. In the latter case, it was hypothesized that the cytoplasmic tail or the transmembrane domain was needed to interact with the viral matrix 1 (M1) protein to facilitate virus assembly (77). However, this hypothesis was not addressed experimentally. Although we did not evaluate whether type A or B virus HA proteins have similar binding affinities for influenza A virus M1, a sufficient interaction presumably takes place to provide comparable multicycle growth and sufficient HA pseudotyping in vitro (Fig. 2 and 3; Table 1). Interestingly, these protein domains overlap the packaging signals of the HA segment, where the signal peptide (51 nt) constitutes the 3′ packaging signal and the transmembrane domain and cytoplasmic tail (108 nt) coincide with the 5′ packaging signal (28, 63). Thus, instead of reconstituting protein-protein interactions in the chimeric influenza A/B viruses, the authors likely restored the compatibility of packaging signals for the incorporation of specific segments into budding virions.
To address if the different promoter signals found at the NCRs of segment termini account for the lack of intertypic reassortment or the ability to generate recombinant intertypic viruses, we performed minigenome assays where a viral RNA-like reporter gene was driven by the influenza A or B virus NCR promoter. Our results indicate that the RdRp of influenza A or B virus can replicate and transcribe reporter genes flanked by heterotypic NCRs, suggesting that in coinfected cells, gene segments would be replicated by both viral RdRps (Fig. 4) (21, 78). However, our and previous results indicate that the NP of influenza B virus inhibits type A RdRp activity (56). This mechanism could clearly account for the lack of segment reassortment between influenza A and B viruses but does not explain why influenza A viruses carrying influenza B virus segments in the absence of influenza B virus NP cannot be generated.
Our observations left the possibility that inefficient gene incorporation restricts influenza A and B virus intertypic reassortment. Influenza A virus RNA molecules direct the specific incorporation of the eight unique vRNA segments into budding virions using packaging signals, which consist of the NCRs and nucleotides localized to the coding termini of each segment (27, 28, 5767). Influenza B virus likely packages genomic segments in a manner similar to that used by influenza A virus, because the nucleotides minimally required for influenza B virus HA packaging are also within the coding termini (Fig. 5). Thus, in cells coinfected with influenza A and B viruses, the packaging signals either are incompatible or have distinct interactions that prevent intertypic packaging events. However, we have shown that appending the influenza A virus packaging signals to influenza B virus segments bypassed this regulation for both HA and NA (Fig. 6). The mechanism by which packaging signals exert their function is not yet known, but detailed supramolecular interaction networks have been proposed on the basis of cryo-electron microscopy, in vitro RNA-RNA interactions, and the generation of recombinant viruses (7981). Additionally, the authors of these studies were able to demonstrate that in the context of H3N2 and H5N2, certain segments preferentially reassort together, perhaps due to the RNA interactions found within packaging signals. Thus, it is plausible that if RNA-RNA interactions are indeed required for segment-specific packaging, then the chimeric segments that we have described herein should interact with their cognate H1N1 segments.
Current methodologies to produce influenza vaccine seed rely on either reverse genetics (31) or classic virus reassortment (82), where seasonal strains that contain the desired HA and NA are forced to reassort with a virus that has high rates of growth in vitro. Our results demonstrate that viable chimeric influenza A/B viruses were immunogenic in mice (Fig. 7; Table 2). Additionally, our ability to reassort chimeric influenza A/B virus with currently circulating pH1N1 suggests that the appended packaging signals are sufficient to drive incorporation into new backbones (Fig. 8). The finding that an influenza A virus containing influenza B virus HA and NA not only was immunogenic but also grew well in vitro suggests that it is possible to develop a single-backbone vaccine platform where the virus backbone provides a high-yield phenotype and circulating influenza A or B virus HA and NA are incorporated via packaging signals. However, further research on its efficacy as an inactivated vaccine and the mutations that provide an attenuated phenotype for a live attenuated vaccine remains to be conducted.

ACKNOWLEDGMENTS

We thank Ron A. M. Fouchier (Erasmus Medical Center) for the pHW2000 Flu-GFP plasmid, Thomas M. Moran at the Center for Therapeutic Antibody Discovery at the Icahn School of Medicine at Mount Sinai for the HT103 and 15B6 monoclonal antibodies, Peter Palese and Adolfo García-Sastre (Icahn School of Medicine at Mount Sinai) for reagents, B. Paige Lawrence (University of Rochester) for the X31 virus, and John J. Treanor (University of Rochester) for anti-B/Vic and anti-B/Yam polyclonal sheep antisera. We also thank the NIAID Biodefense and Emerging Infectious Research Resources Repository (BEI Resources) for providing antibodies NR-3114, NR-3165, and NR-4540.
S.F.B. is currently supported by a University of Rochester immunology training grant (AI 007285-26). A.N. is the recipient of the University of Rochester Vaccine Fellowship (2013) and a Centers for Excellence in Influenza Research & Surveillance (CEIRS) intercollaborative training grant (2013). W.D. is currently supported by a University of Rochester pulmonary training grant (T32-HL66988 12). Research in the L.M.-S. laboratory is funded by NIH grants RO1 AI077719 and R03AI099681-01A1, the NIAID Centers of Excellence for Influenza Research and Surveillance (CEIRS HHSN266200700008C), and the University of Rochester Center for Biodefense Immune Modeling (CBIM HHSN272201000055C).

REFERENCES

1.
Palese P and Shaw ML. 2007. Orthomyxoviridae: the viruses and their replication. In Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA, Roizman B, and Straus SE (ed), Fields virology, 5th ed. Lippincott Williams & Wilkins, Philadelphia, PA.
2.
Chen R and Holmes EC. 2008. The evolutionary dynamics of human influenza B virus. J. Mol. Evol. 66:655–663.
3.
Osterhaus AD, Rimmelzwaan GF, Martina BE, Bestebroer TM, and Fouchier RA. 2000. Influenza B virus in seals. Science 288:1051–1053.
4.
Baker SF, Guo H, Albrecht RA, Garcia-Sastre A, Topham DJ, and Martinez-Sobrido L. 2013. Protection against lethal influenza with a viral mimic. J. Virol. 87:8591–8605.
5.
Jackson D, Elderfield RA, and Barclay WS. 2011. Molecular studies of influenza B virus in the reverse genetics era. J. Gen. Virol. 92:1–17.
6.
Schild GC, Oxford JS, de Jong JC, and Webster RG. 1983. Evidence for host-cell selection of influenza virus antigenic variants. Nature 303:706–709.
7.
Tong S, Li Y, Rivailler P, Conrardy C, Castillo DA, Chen LM, Recuenco S, Ellison JA, Davis CT, York IA, Turmelle AS, Moran D, Rogers S, Shi M, Tao Y, Weil MR, Tang K, Rowe LA, Sammons S, Xu X, Frace M, Lindblade KA, Cox NJ, Anderson LJ, Rupprecht CE, and Donis RO. 2012. A distinct lineage of influenza A virus from bats. Proc. Natl. Acad. Sci. U. S. A. 109:4269–4274.
8.
Tong S, Zhu X, Li Y, Shi M, Zhang J, Bourgeois M, Yang H, Chen X, Recuenco S, Gomez J, Chen LM, Johnson A, Tao Y, Dreyfus C, Yu W, McBride R, Carney PJ, Gilbert AT, Chang J, Guo Z, Davis CT, Paulson JC, Stevens J, Rupprecht CE, Holmes EC, Wilson IA, and Donis RO. 2013. New World bats harbor diverse influenza A viruses. PLoS Pathog. 9:e1003657.
9.
McCullers JA, Saito T, and Iverson AR. 2004. Multiple genotypes of influenza B virus circulated between 1979 and 2003. J. Virol. 78:12817–12828.
10.
Rota P, Wallis TR, Harmon MW, Rota JS, Kendal AP, and Nerome K. 1990. Cocirculation of two distinct evolutionary lineages of influenza type B virus since 1983. Virology 175:59–68.
11.
Kanegae Y, Sugita S, Endo A, Ishida M, Senya S, Osako K, Nerome K, and Oya A. 1990. Evolutionary pattern of the hemagglutinin gene of influenza B viruses isolated in Japan: cocirculating lineages in the same epidemic season. J. Virol. 64:2860–2865.
12.
McCullers JA, Wang GC, He S, and Webster RG. 1999. Reassortment and insertion-deletion are strategies for the evolution of influenza B viruses in nature. J. Virol. 73:7343–7348.
13.
Xu X, Lindstrom SE, Shaw MW, Smith CB, Hall HE, Mungall BA, Subbarao K, Cox NJ, and Klimov A. 2004. Reassortment and evolution of current human influenza A and B viruses. Virus Res. 103:55–60.
14.
Mikheeva A and Ghendon YZ. 1982. Intrinsic interference between influenza A and B viruses. Arch. Virol. 73:287–294.
15.
Kaverin NV, Varich NL, Sklyanskaya EI, Amvrosieva TV, Petrik J, and Vovk TC. 1983. Studies on heterotypic interference between influenza A and B viruses: a differential inhibition of the synthesis of viral proteins and RNAs. J. Gen. Virol. 64(Pt 10):2139–2146.
16.
Tobita K, Tanaka T, Goto H, and Feng SY. 1983. Temperature-sensitive influenza A virus clones originated by a cross between A/Aichi/2/68 (H3N2) and B/Yamagata/1/73. Arch. Virol. 75:17–27.
17.
Ghate AA and Air GM. 1999. Influenza type B neuraminidase can replace the function of type A neuraminidase. Virology 264:265–277.
18.
Flandorfer A, Garcia-Sastre A, Basler CF, and Palese P. 2003. Chimeric influenza A viruses with a functional influenza B virus neuraminidase or hemagglutinin. J. Virol. 77:9116–9123.
19.
Horimoto T, Takada A, Iwatsuki-Horimoto K, Hatta M, Goto H, and Kawaoka Y. 2003. Generation of influenza A viruses with chimeric (type A/B) hemagglutinins. J. Virol. 77:8031–8038.
20.
Muster T, Subbarao EK, Enami M, Murphy BR, and Palese P. 1991. An influenza A virus containing influenza B virus 5′ and 3′ noncoding regions on the neuraminidase gene is attenuated in mice. Proc. Natl. Acad. Sci. U. S. A. 88:5177–5181.
21.
Crescenzo-Chaigne B, Naffakh N, and van der Werf S. 1999. Comparative analysis of the ability of the polymerase complexes of influenza viruses type A, B and C to assemble into functional RNPs that allow expression and replication of heterotypic model RNA templates in vivo. Virology 265:342–353.
22.
Iwatsuki-Horimoto K, Hatta Y, Hatta M, Muramoto Y, Chen H, Kawaoka Y, and Horimoto T. 2008. Limited compatibility between the RNA polymerase components of influenza virus type A and B. Virus Res. 135:161–165.
23.
Fodor E, Pritlove DC, and Brownlee GG. 1994. The influenza virus panhandle is involved in the initiation of transcription. J. Virol. 68:4092–4096.
24.
Stoeckle MY, Shaw MW, and Choppin PW. 1987. Segment-specific and common nucleotide sequences in the noncoding regions of influenza B virus genome RNAs. Proc. Natl. Acad. Sci. U. S. A. 84:2703–2707.
25.
Hutchinson EC, von Kirchbach JC, Gog JR, and Digard P. 2010. Genome packaging in influenza A virus. J. Gen. Virol. 91:313–328.
26.
Martinez-Sobrido L, Cadagan R, Steel J, Basler CF, Palese P, Moran TM, and Garcia-Sastre A. 2010. Hemagglutinin-pseudotyped green fluorescent protein-expressing influenza viruses for the detection of influenza virus neutralizing antibodies. J. Virol. 84:2157–2163.
27.
Fujii Y, Goto H, Watanabe T, Yoshida T, and Kawaoka Y. 2003. Selective incorporation of influenza virus RNA segments into virions. Proc. Natl. Acad. Sci. U. S. A. 100:2002–2007.
28.
Marsh GA, Hatami R, and Palese P. 2007. Specific residues of the influenza A virus hemagglutinin viral RNA are important for efficient packaging into budding virions. J. Virol. 81:9727–9736.
29.
Rodrigo WW, de la Torre JC, and Martinez-Sobrido L. 2011. Use of single-cycle infectious lymphocytic choriomeningitis virus to study hemorrhagic fever arenaviruses. J. Virol. 85:1684–1695.
30.
Martinez-Sobrido L and Garcia-Sastre A. 2010. Generation of recombinant influenza virus from plasmid DNA. J. Vis. Exp. 3:2057.
31.
Schickli JH, Flandorfer A, Nakaya T, Martinez-Sobrido L, Garcia-Sastre A, and Palese P. 2001. Plasmid-only rescue of influenza A virus vaccine candidates. Philos. Trans. R. Soc. Lond. B Biol. Sci. 356:1965–1973.
32.
Hai R, Martinez-Sobrido L, Fraser KA, Ayllon J, Garcia-Sastre A, and Palese P. 2008. Influenza B virus NS1-truncated mutants: live-attenuated vaccine approach. J. Virol. 82:10580–10590.
33.
Hai R, Schmolke M, Varga ZT, Manicassamy B, Wang TT, Belser JA, Pearce MB, Garcia-Sastre A, Tumpey TM, and Palese P. 2010. PB1-F2 expression by the 2009 pandemic H1N1 influenza virus has minimal impact on virulence in animal models. J. Virol. 84:4442–4450.
34.
Hoffmann E, Neumann G, Hobom G, Webster RG, and Kawaoka Y. 2000. “Ambisense” approach for the generation of influenza A virus: vRNA and mRNA synthesis from one template. Virology 267:310–317.
35.
Fodor E, Devenish L, Engelhardt OG, Palese P, Brownlee GG, and Garcia-Sastre A. 1999. Rescue of influenza A virus from recombinant DNA. J. Virol. 73:9679–9682.
36.
Kilbourne ED. 1969. Future influenza vaccines and the use of genetic recombinants. Bull. World Health Organ. 41:643–645.
37.
Pena L, Sutton T, Chockalingam A, Kumar S, Angel M, Shao H, Chen H, Li W, and Perez DR. 2013. Influenza viruses with rearranged genomes as live-attenuated vaccines. J. Virol. 87:5118–5127.
38.
Chapman TJ, Castrucci MR, Padrick RC, Bradley LM, and Topham DJ. 2005. Antigen-specific and non-specific CD4+ T cell recruitment and proliferation during influenza infection. Virology 340:296–306.
39.
O'Neill RE, Talon J, and Palese P. 1998. The influenza virus NEP (NS2 protein) mediates the nuclear export of viral ribonucleoproteins. EMBO J. 17:288–296.
40.
Bauman JD, Patel D, Baker SF, Vijayan RS, Xiang A, Parhi AK, Martinez-Sobrido L, LaVoie EJ, Das K, and Arnold E. 2013. Crystallographic fragment screening and structure-based optimization yields a new class of influenza endonuclease inhibitors. ACS Chem. Biol. 8:2501–2508.
41.
Hoffmann E, Stech J, Guan Y, Webster RG, and Perez DR. 2001. Universal primer set for the full-length amplification of all influenza A viruses. Arch. Virol. 146:2275–2289.
42.
Hoffmann E, Neumann G, Kawaoka Y, Hobom G, and Webster RG. 2000. A DNA transfection system for generation of influenza A virus from eight plasmids. Proc. Natl. Acad. Sci. U. S. A. 97:6108–6113.
43.
Song H, Nieto GR, and Perez DR. 2007. A new generation of modified live-attenuated avian influenza viruses using a two-strategy combination as potential vaccine candidates. J. Virol. 81:9238–9248.
44.
Hoffmann E, Mahmood K, Yang CF, Webster RG, Greenberg HB, and Kemble G. 2002. Rescue of influenza B virus from eight plasmids. Proc. Natl. Acad. Sci. U. S. A. 99:11411–11416.
45.
Mayer D, Molawi K, Martinez-Sobrido L, Ghanem A, Thomas S, Baginsky S, Grossmann J, Garcia-Sastre A, and Schwemmle M. 2007. Identification of cellular interaction partners of the influenza virus ribonucleoprotein complex and polymerase complex using proteomic-based approaches. J. Proteome Res. 6:672–682.
46.
Martinez-Sobrido L, Zuniga EI, Rosario D, Garcia-Sastre A, and de la Torre JC. 2006. Inhibition of the type I interferon response by the nucleoprotein of the prototypic arenavirus lymphocytic choriomeningitis virus. J. Virol. 80:9192–9199.
47.
Rimmelzwaan GF, Nieuwkoop NJ, de Mutsert G, Boon AC, Kuiken T, Fouchier RA, and Osterhaus AD. 2007. Attachment of infectious influenza A viruses of various subtypes to live mammalian and avian cells as measured by flow cytometry. Virus Res. 129:175–181.
48.
Medina RA, Manicassamy B, Stertz S, Seibert CW, Hai R, Belshe RB, Frey SE, Basler CF, Palese P, and Garcia-Sastre A. 2010. Pandemic 2009 H1N1 vaccine protects against 1918 Spanish influenza virus. Nat. Commun. 1:28.
49.
Reale MA, Manheimer AJ, Moran TM, Norton G, Bona CA, and Schulman JL. 1986. Characterization of monoclonal antibodies specific for sequential influenza A/PR/8/34 virus variants. J. Immunol. 137:1352–1358.
50.
Ortiz-Riano E, Cheng BY, de la Torre JC, and Martinez-Sobrido L. 2011. The C-terminal region of lymphocytic choriomeningitis virus nucleoprotein contains distinct and segregable functional domains involved in NP-Z interaction and counteraction of the type I interferon response. J. Virol. 85:13038–13048.
51.
Guo H, Santiago F, Lambert K, Takimoto T, and Topham DJ. 2011. T cell-mediated protection against lethal 2009 pandemic H1N1 influenza virus infection in a mouse model. J. Virol. 85:448–455.
52.
Mibayashi M, Martinez-Sobrido L, Loo YM, Cardenas WB, Gale M Jr, and Garcia-Sastre A. 2007. Inhibition of retinoic acid-inducible gene I-mediated induction of beta interferon by the NS1 protein of influenza A virus. J. Virol. 81:514–524.
53.
National Research Council. 2011. Guide for the care and use of laboratory animals, 8th ed. National Academies Press, Washington, DC.
54.
Reed LJ and Muench H. 1938. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27:493–497.
55.
Garten RJ, Davis CT, Russell CA, Shu B, Lindstrom S, Balish A, Sessions WM, Xu X, Skepner E, Deyde V, Okomo-Adhiambo M, Gubareva L, Barnes J, Smith CB, Emery SL, Hillman MJ, Rivailler P, Smagala J, de Graaf M, Burke DF, Fouchier RA, Pappas C, Alpuche-Aranda CM, Lopez-Gatell H, Olivera H, Lopez I, Myers CA, Faix D, Blair PJ, Yu C, Keene KM, Dotson PD Jr, Boxrud D, Sambol AR, Abid SH, St George K, Bannerman T, Moore AL, Stringer DJ, Blevins P, Demmler-Harrison GJ, Ginsberg M, Kriner P, Waterman S, Smole S, Guevara HF, Belongia EA, Clark PA, Beatrice ST, et al. 2009. Antigenic and genetic characteristics of swine-origin 2009 A(H1N1) influenza viruses circulating in humans. Science 325:197–201.
56.
Wanitchang A, Narkpuk J, Jaru-ampornpan P, Jengarn J, and Jongkaewwattana A. 2012. Inhibition of influenza A virus replication by influenza B virus nucleoprotein: an insight into interference between influenza A and B viruses. Virology 432:194–203.
57.
Dos Santos Afonso E, Escriou N, Leclercq I, van der Werf S, and Naffakh N. 2005. The generation of recombinant influenza A viruses expressing a PB2 fusion protein requires the conservation of a packaging signal overlapping the coding and noncoding regions at the 5′ end of the PB2 segment. Virology 341:34–46.
58.
Fujii K, Fujii Y, Noda T, Muramoto Y, Watanabe T, Takada A, Goto H, Horimoto T, and Kawaoka Y. 2005. Importance of both the coding and the segment-specific noncoding regions of the influenza A virus NS segment for its efficient incorporation into virions. J. Virol. 79:3766–3774.
59.
Fujii K, Ozawa M, Iwatsuki-Horimoto K, Horimoto T, and Kawaoka Y. 2009. Incorporation of influenza A virus genome segments does not absolutely require wild-type sequences. J. Gen. Virol. 90:1734–1740.
60.
Gog JR, Afonso Edos S, Dalton RM, Leclercq I, Tiley L, Elton D, von Kirchbach JC, Naffakh N, Escriou N, and Digard P. 2007. Codon conservation in the influenza A virus genome defines RNA packaging signals. Nucleic Acids Res. 35:1897–1907.
61.
Liang Y, Hong Y, and Parslow TG. 2005. cis-Acting packaging signals in the influenza virus PB1, PB2, and PA genomic RNA segments. J. Virol. 79:10348–10355.
62.
Liang Y, Huang T, Ly H, Parslow TG, and Liang Y. 2008. Mutational analyses of packaging signals in influenza virus PA, PB1, and PB2 genomic RNA segments. J. Virol. 82:229–236.
63.
Marsh GA, Rabadan R, Levine AJ, and Palese P. 2008. Highly conserved regions of influenza A virus polymerase gene segments are critical for efficient viral RNA packaging. J. Virol. 82:2295–2304.
64.
Muramoto Y, Takada A, Fujii K, Noda T, Iwatsuki-Horimoto K, Watanabe S, Horimoto T, Kida H, and Kawaoka Y. 2006. Hierarchy among viral RNA (vRNA) segments in their role in vRNA incorporation into influenza A virions. J. Virol. 80:2318–2325.
65.
Ozawa M, Fujii K, Muramoto Y, Yamada S, Yamayoshi S, Takada A, Goto H, Horimoto T, and Kawaoka Y. 2007. Contributions of two nuclear localization signals of influenza A virus nucleoprotein to viral replication. J. Virol. 81:30–41.
66.
Ozawa M, Maeda J, Iwatsuki-Horimoto K, Watanabe S, Goto H, Horimoto T, and Kawaoka Y. 2009. Nucleotide sequence requirements at the 5′ end of the influenza A virus M RNA segment for efficient virus replication. J. Virol. 83:3384–3388.
67.
Watanabe T, Watanabe S, Noda T, Fujii Y, and Kawaoka Y. 2003. Exploitation of nucleic acid packaging signals to generate a novel influenza virus-based vector stably expressing two foreign genes. J. Virol. 77:10575–10583.
68.
Pica N, Iyer A, Ramos I, Bouvier NM, Fernandez-Sesma A, Garcia-Sastre A, Lowen AC, Palese P, and Steel J. 2011. The DBA.2 mouse is susceptible to disease following infection with a broad, but limited, range of influenza A and B viruses. J. Virol. 85:12825–12829.
69.
Kobasa D, Takada A, Shinya K, Hatta M, Halfmann P, Theriault S, Suzuki H, Nishimura H, Mitamura K, Sugaya N, Usui T, Murata T, Maeda Y, Watanabe S, Suresh M, Suzuki T, Suzuki Y, Feldmann H, and Kawaoka Y. 2004. Enhanced virulence of influenza A viruses with the haemagglutinin of the 1918 pandemic virus. Nature 431:703–707.
70.
Carragher DM, Kaminski DA, Moquin A, Hartson L, and Randall TD. 2008. A novel role for non-neutralizing antibodies against nucleoprotein in facilitating resistance to influenza virus. J. Immunol. 181:4168–4176.
71.
Dowdle WN, Kendal AP, and Noble GR. 1979. Influenza viruses, p 585–609. In Lennette EH and Schmidt NJ (ed), Diagnostic procedures for viral, rickettsial, and chlamydial infections, 5th ed. American Public Health Association, Washington, DC.
72.
Jin H, Lu B, Zhou H, Ma C, Zhao J, Yang CF, Kemble G, and Greenberg H. 2003. Multiple amino acid residues confer temperature sensitivity to human influenza virus vaccine strains (FluMist) derived from cold-adapted A/Ann Arbor/6/60. Virology 306:18–24.
73.
Yewdell JW, Bennink JR, and Hosaka Y. 1988. Cells process exogenous proteins for recognition by cytotoxic T lymphocytes. Science 239:637–640.
74.
Suzuki Y and Nei M. 2002. Origin and evolution of influenza virus hemagglutinin genes. Mol. Biol. Evol. 19:501–509.
75.
Taubenberger JK and Kash JC. 2010. Influenza virus evolution, host adaptation, and pandemic formation. Cell Host Microbe 7:440–451.
76.
Hai R, Garcia-Sastre A, Swayne DE, and Palese P. 2011. A reassortment-incompetent live attenuated influenza virus vaccine for protection against pandemic virus strains. J. Virol. 85:6832–6843.
77.
Ali A, Avalos RT, Ponimaskin E, and Nayak DP. 2000. Influenza virus assembly: effect of influenza virus glycoproteins on the membrane association of M1 protein. J. Virol. 74:8709–8719.
78.
Jambrina E, Barcena J, Uez O, and Portela A. 1997. The three subunits of the polymerase and the nucleoprotein of influenza B virus are the minimum set of viral proteins required for expression of a model RNA template. Virology 235:209–217.
79.
Fournier E, Moules V, Essere B, Paillart JC, Sirbat JD, Isel C, Cavalier A, Rolland JP, Thomas D, Lina B, and Marquet R. 2012. A supramolecular assembly formed by influenza A virus genomic RNA segments. Nucleic Acids Res. 40:2197–2209.
80.
Essere B, Yver M, Gavazzi C, Terrier O, Isel C, Fournier E, Giroux F, Textoris J, Julien T, Socratous C, Rosa-Calatrava M, Lina B, Marquet R, and Moules V. 2013. Critical role of segment-specific packaging signals in genetic reassortment of influenza A viruses. Proc. Natl. Acad. Sci. U. S. A. 110:E3840–E3848.
81.
Gavazzi C, Isel C, Fournier E, Moules V, Cavalier A, Thomas D, Lina B, and Marquet R. 2013. An in vitro network of intermolecular interactions between viral RNA segments of an avian H5N2 influenza A virus: comparison with a human H3N2 virus. Nucleic Acids Res. 41:1241–1254.
82.
Kilbourne ED, Schulman JL, Schild GC, Schloer G, Swanson J, and Bucher D. 1971. Related studies of a recombinant influenza-virus vaccine. I. Derivation and characterization of virus and vaccine. J. Infect. Dis. 124:449–462.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 88Number 1815 September 2014
Pages: 10778 - 10791
Editor: D. S. Lyles
PubMed: 25008914

History

Received: 19 May 2014
Accepted: 30 June 2014
Published online: 15 September 2014

Permissions

Request permissions for this article.

Contributors

Authors

Steven F. Baker
Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
Aitor Nogales
Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
Courtney Finch
Department of Veterinary Medicine, University of Maryland College Park, and Virginia-Maryland Regional College of Veterinary Medicine, College Park, Maryland, USA
Kevin M. Tuffy
Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
William Domm
Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
Daniel R. Perez
Department of Veterinary Medicine, University of Maryland College Park, and Virginia-Maryland Regional College of Veterinary Medicine, College Park, Maryland, USA
David J. Topham
Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
New York Influenza Center of Excellence, University of Rochester, Rochester, New York, USA
David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, New York, USA
Luis Martínez-Sobrido
Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA

Editor

D. S. Lyles
Editor

Notes

Address correspondence to Luis Martínez-Sobrido, [email protected].

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy