Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Journal of Virology
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Vaccines and Antiviral Agents

Temperature-Sensitive Live-Attenuated Canine Influenza Virus H3N8 Vaccine

Aitor Nogales, Laura Rodriguez, Caroline Chauché, Kai Huang, Emma C. Reilly, David J. Topham, Pablo R. Murcia, Colin R. Parrish, Luis Martínez-Sobrido
Terence S. Dermody, Editor
Aitor Nogales
aDepartment of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laura Rodriguez
aDepartment of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Caroline Chauché
bMRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kai Huang
cBaker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Emma C. Reilly
dDavid H. Smith Center for Vaccine Biology and Immunology and Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David J. Topham
dDavid H. Smith Center for Vaccine Biology and Immunology and Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for David J. Topham
Pablo R. Murcia
bMRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Pablo R. Murcia
Colin R. Parrish
cBaker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Colin R. Parrish
Luis Martínez-Sobrido
aDepartment of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Terence S. Dermody
University of Pittsburgh School of Medicine
Roles: Editor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/JVI.02211-16
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Canine influenza is a respiratory disease of dogs caused by canine influenza virus (CIV). CIV subtypes responsible for influenza in dogs include H3N8, which originated from the transfer of H3N8 equine influenza virus to dogs; and the H3N2 CIV, which is an avian-origin virus that adapted to infect dogs. Influenza infections are most effectively prevented through vaccination to reduce transmission and future infection. Currently, only inactivated influenza vaccines (IIVs) are available for the prevention of CIV in dogs. However, the efficacy of IIVs is suboptimal, and novel approaches are necessary for the prevention of disease caused by this canine respiratory pathogen. Using reverse genetics techniques, we have developed a live-attenuated CIV vaccine (LACIV) for the prevention of H3N8 CIV. The H3N8 LACIV replicates efficiently in canine cells at 33°C but is impaired at temperatures of 37 to 39°C and was attenuated compared to wild-type H3N8 CIV in vivo and ex vivo. The LACIV was able to induce protection against H3N8 CIV challenge with a single intranasal inoculation in mice. Immunogenicity and protection efficacy were better than that observed with a commercial CIV H3N8 IIV but provided limited cross-reactive immunity and heterologous protection against H3N2 CIV. These results demonstrate the feasibility of implementing a LAIV approach for the prevention and control of H3N8 CIV in dogs and suggest the need for a new LAIV for the control of H3N2 CIV.

IMPORTANCE Two influenza A virus subtypes has been reported in dogs in the last 16 years: the canine influenza viruses (CIV) H3N8 and H3N2 of equine and avian origins, respectively. To date, only inactivated influenza vaccines (IIVs) are available to prevent CIV infections. Here, we report the generation of a recombinant, temperature-sensitive H3N8 CIV as a live-attenuated influenza vaccine (LAIV), which was attenuated in mice and dog tracheal, explants compared to CIV H3N8 wild type. A single dose of H3N8 LACIV showed immunogenicity and protection against a homologous challenge that was better than that conferred with an H3N8 IIV, demonstrating the feasibility of implementing a LAIV approach for the improved control of H3N8 CIV infections in dogs.

INTRODUCTION

Influenza A viruses (IAVs) are enveloped viruses that belong to the Orthomyxoviridae family and contain a genome that comprises eight single-stranded negative-sense RNA segments that encode 10 to 14 proteins (1). The hemagglutinin (HA) and the neuraminidase (NA) glycoproteins are the major antigenic determinants of IAV and are essential for receptor binding and fusion and virion release, respectively (2). IAV HA and NA glycoproteins within infected organisms and populations are driven to evolve antigenic variants via immunological pressure and in humans and some other hosts positive selection of viruses occurs gradually in a process known as antigenic drift (3). The antigenic diversity of glycoproteins is used to further classify IAVs, of which there are 18 HA and 11 NA subtypes (4, 5). In addition, antigenically distinct isolates can also exist within the same subtype, referred to as drifted variants. IAVs exist mainly in the wild aquatic fowl reservoir (6–9), and only a small number of mammalian hosts are currently recognized as sustaining transmission of IAVs.

Canine influenza is a contagious respiratory disease of dogs caused by two IAVs: the H3N8 equine-origin influenza virus that transferred to dogs in the United States around 1999 (10) and the avian virus-like H3N2 that transferred to dogs in Asia around 2005 (11). In 2015 an outbreak of H3N2 canine influenza virus (CIV) occurred in the United States that was due to a virus similar to those detected in dogs in Asia (12). The H3N2 CIV has also been isolated from cats in a shelter in South Korea (13, 14). These CIVs represent new threats to canine health in the United States and worldwide, since the virus may be spread through the racing track circuit, as was the case of the H3N8 strain (10, 15), while both viruses are spread widely within and among animal shelters and kennels (10, 16, 17). CIVs are still relatively new viruses and because of the low levels of infection and immunity among the broader population most dogs are susceptible to infection. Most dogs infected by CIVs show only a mild respiratory illness, but severe outcomes are also observed (18).

The recent emergence of CIVs (H3N8 and H3N2 CIVs) has increased the host range of IAVs. The continuous circulation of CIVs in dog populations creates opportunities for exposure of humans and other animals. Since dogs are susceptible to mammalian (equine-origin H3N8 CIV) and avian (avian-origin H3N2 CIV) IAVs, they may have the potential to act as “mixing vessel” hosts for new IAV strains with potential for human infection. Reassortments between H3N2 CIVs and human pandemic H1N1 IAV have been reported (19, 20), and the introduction of novel, antigenically distinct glycoproteins (HA and NA) into the backbones of human IAVs have been associated with human pandemics (21).

Vaccination is accepted as an effective strategy for the prevention of influenza infections (22, 23). To date, three types of influenza virus vaccines have been approved by the U.S. Food and Drug Administration for human use: recombinant viral HAs, inactivated influenza vaccines (IIVs), and live-attenuated influenza vaccines (LAIVs) (22, 24–27). In dogs, only IIV against both H3N8 and H3N2 CIVs are commercially available. However, we have recently reported the generation of recombinant H3N8 CIVs containing truncated or a deleted nonstructural 1 (NS1) protein as potential LAIVs candidates for the treatment of H3N8 CIV infections (28).

IIVs are administered intramuscularly and elicit humoral immunity by inducing the production of neutralizing antibodies that target epitopes on HA (26, 29). On the other hand, LAIVs more closely mimic the natural route of viral infection and elicit both cellular and humoral immune responses (24), providing better immunogenicity and protection (22, 28, 30).

In mammals IAV is a respiratory pathogen that replicates in the cooler (33°C) upper respiratory tract, in addition to replicating in the warmer (37°C) conditions of the lower respiratory tract (31). This temperature difference has allowed for the development of cold-adapted (ca), temperature-sensitive (ts), attenuated (att) viruses that replicate in the upper respiratory tract but do not damage the lower respiratory tract due to the elevated temperatures restricting replication (32). For human viruses these ca, ts, and att properties have been mapped to five amino acid residues located in three viral proteins of A/Ann Arbor/6/60 H2N2 (A/AA/6/60): polymerase basic 2 (PB2) N265S; polymerase basic 1 (PB1) K391E, D581G, and A661T; and nucleoprotein (NP) D34G (33, 34). The mechanisms of attenuation are not fully understood but most likely involve multiple steps in the replication cycle of the virus (32). Importantly, when the ts signature of A/AA/6/60 was introduced into influenza A/Puerto Rico/8/34 H1N1 (PR8) or A/California/04/09 H1N1 (pH1N1) viruses, a similar ts phenotype was observed in tissue culture cells and in the mouse model of infection (35–37).

In order to develop a LAIV for the treatment of CIV H3N8 infections, we introduced the ts, ca, att mutations identified in the A/AA/6/60 LAIV into CIV H3N8 (referred to henceforth as LACIV H3N8) using reverse genetics (38). LACIV H3N8 replicated efficiently in vitro at 33°C but not at 37 or 39°C. Compared to CIV H3N8 wild-type (WT), LACIV H3N8 was attenuated ex vivo and in vivo but was able to induce protective immunity in mice against H3N8 WT upon a single intranasal (i.n.) dose, demonstrating its feasibility as a safe, immunogenic, and protective LAIV candidate.

RESULTS

Generation and characterization of H3N8 LACIV.We introduced four ts mutations identified in previous studies into the PB2 and PB1 genes of H3N8 CIV (33, 39) (Fig. 1). No mutation was introduced into the viral NP since H3N8 CIV NP already contains a G at position 34.

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

Effect of temperature on the polymerase activity of H3N8 LACIV. (A) Schematic representation of segments 1 (PB2) and 2 (PB1) of WT (black) and LACIV (gray) H3N8 CIV. Amino acid substitutions N265S (PB2) and K391E, E581G, and A661T (PB1) to generate the H3N8 LACIV are indicated. (B) Minigenome activity. MDCK cells (12-well plate format, 3 × 105 cells/well, triplicates) were transiently cotransfected with 250 ng of ambisense pDZ expression plasmids encoding the minimal requirements for viral genome replication and gene transcription (PB2, PB1, PA, and NP), together with 500 ng of a vRNA-like expression plasmid encoding Gaussia luciferase (Gluc) under the control of the canine polymerase I promoter (cpPol-I Gluc), and 100 ng of a pCAGGS Cypridina luciferase (Cluc) plasmid to normalize transfection efficiencies. After transfection, cells were placed at 33, 37, or 39°C, and viral replication and transcription was evaluated 24 h later by luminescence (Gluc). Gluc activity was normalized to that of Cluc. The data represent means and SD. Normalized reporter expression is relative to that in the absence of pDZ NP plasmid. Data were represented as relative activity considering WT H3N8 polymerase activity at each temperature as 100%. *, P < 0.05 (Student t test).

To determine whether the mutations introduced into the PB2 and PB1 genes confer a ts phenotype to the H3N8 CIV polymerase, we performed a minigenome assay. Both WT and LACIV H3N8 resulted in similar Gaussia luciferase (Gluc) expression levels at 33°C (Fig. 1B), but Gluc expression was reduced at higher temperatures (37 and 39°C) in cells transfected with the H3N8 LACIV plasmids. This shows that those mutations resulted in a ts phenotype when introduced in the H3N8 CIV, as previously described for A/AA/6/60 H2N2 and other influenza viruses (35–37).

We next generated an H3N8 LACIV using plasmid-based reverse genetic approaches (40, 41) and evaluated the viral replication kinetics in MDCK cells infected at a low (0.001) multiplicity of infection (MOI) and compared them to the WT H3N8 CIV (Fig. 2). At 33°C, both WT and LACIV H3N8 grew with the same kinetics and reached similar high titers (107 FFU/ml) at 48 to 72 h postinfection (p.i.) (Fig. 2A). At higher temperatures (37 and 39°C), the WT H3N8 CIV replicated at levels similar to those observed at 33°C, while H3N8 LACIV titers were reduced ca. 2 to 3 logs at 37°C (Fig. 2B) or was not detected at 39°C (Fig. 2C). These results demonstrate that mutations introduced in PB2 and PB1 conferred a ts phenotype to H3N8 CIV.

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Characterization of H3N8 LACIV in vitro: MDCK cells (12-well plate format, 3 × 105 cells/well, triplicates) were infected (MOI of 0.001) with WT (black diamonds) and LACIV (gray squares) H3N8 CIVs and incubated at 33°C (A), 37°C (B), and 39°C (C). TCS were collected at 12, 24, 48, 72, and 96 h p.i., and the viral titers were determined by immunofocus assay (FFU/ml). Data represent the means and SD of the results determined in triplicate. Dotted lines indicate the limit of detection (200 FFU/ml). *, P < 0.05 (Student t test).

LACIV H3N8 is attenuated in vivo in mice.Since the H3N8 LACIV presented defects in replication at higher (37 and 39°C) temperatures, we next investigated whether H3N8 LACIV was also attenuated in mice. No signs of infection were detected after infection with WT H3N8 CIV. Therefore, CIV titers in the lungs of infected (105 PFU) mice were determined on days 2 (n = 3) and 4 (n = 3) p.i. and used as a measure of viral attenuation (Fig. 3). Notably, virus titers in the lungs were only detected in mice inoculated with WT H3N8 CIV, and no virus was detected in mice infected with the H3N8 LACIV. These results indicate that H3N8 LACIV is also attenuated in vivo.

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Attenuation of H3N8 LACIV in vivo. Female 5- to-7-week-old C57BL/6 WT mice (n = 6) were infected i.n. with 105 PFU of WT or LACIV H3N8 CIVs. Three mice were sacrificed at days 2 (black) and 4 (gray) p.i., and the lungs were harvested for virus titrations using an immunofocus assay (FFU/ml). Data represent the means and SD. Dotted line indicate limit of detection (200 FFU/ml). ND, virus not detected.

Intranasal vaccination with H3N8 LACIV induces protective immunity against WT H3N8 CIV challenge.To evaluate the immunity generated by the H3N8 LACIV, mice (n = 6) were vaccinated i.n. with 103 PFU of H3N8 WT or LACIV, mock vaccinated with phosphate-buffered saline (PBS), or vaccinated intramuscularly (i.m.) with 100 μl of Nobivac, a commercial IIV against H3N8 CIV (Fig. 4). Humoral immune responses were evaluated in mice sera collected 2 weeks later (Fig. 4A). Total H3N8 CIV antibody responses were characterized by enzyme-linked immunosorbent assay (ELISA) using cell lysates from mock- or H3N8 CIV-infected MDCK cells (41). Mice vaccinated with the H3N8 LACIV elicited high serum IgG titers against parental H3N8 CIV, whereas the antibody titers of mice vaccinated with Nobivac were lower than those in the H3N8 LACIV or WT vaccinated mice (Fig. 4A). In addition, hemagglutination inhibition (HAI) assays were performed to examine the presence of anti-HA neutralizing antibodies on sera from vaccinated mice (Fig. 4B), showing that HAI titers against CIV H3N8 were higher in mice vaccinated with the H3N8 LACIV than those observed in mice vaccinated with the H3N8 IIV.

FIG 4
  • Open in new tab
  • Download powerpoint
FIG 4

Immunogenicity of H3N8 LACIV: Female 5- to-7-week-old C57BL/6 WT mice (n = 6) were vaccinated i.n. with 103 PFU of WT or LACIV H3N8 CIVs. Mice mock (PBS) vaccinated or vaccinated i.m. with 100 μl of an H3N8 CIV IIV (Nobivac) were used as internal controls. (A) Induction of humoral responses. At 14 days postvaccination, mice were bled, and sera were evaluated for the presence of total IgG antibodies against H3N8 CIV proteins using cell extracts of MDCK-infected cells by ELISA. MDCK mock-infected cell extracts were used to evaluate the specificity of the antibody response. OD, optical density. Data represent the means ± the SD of the results for six individual mice. *, Nobivac versus LACIV; **, WT versus LACIV; ***, WT versus Nobivac (P < 0.05 using the Student t test). (B) HAI titers. Sera from immunized mice were evaluated by HAI using four HAU of WT H3N8 CIV and 2-fold serial dilutions of the indicated sera. ND, not detected. *, WT versus LACIV or Nobivac; or *, LACIV versus Nobivac (P < 0.05 using the Student t test).

To further examine the immunogenicity of LACIV, we evaluated whether the virus can induce a localized CD8 T cell response (Fig. 5). To this end, mice (n = 4) were immunized as described above and, at 10 days p.i., lung and spleen samples were collected and single-cell preparations made for flow cytometric analysis (42). Live CD8 T cells were further gated for a H3N8 CIV-specific population (42, 43). The results showed that vaccination with H3N8 LACIV induced elevated levels of NP and PA-specific lung CD8 T cells, similar to those induced by WT virus in both lungs (Fig. 5A) and the spleen (Fig. 5B). Importantly, animals vaccinated with Nobivac did not show a T cell response either in the lung (Fig. 5A) or the spleen (Fig. 5B), highlighting important differences in induced immunity between H3N8 LACIV and the H3N8 IIV.

FIG 5
  • Open in new tab
  • Download powerpoint
FIG 5

CD8 T cell response induced by H3N8 LACIV. Female 5- to-7-week-old C57BL/6 WT mice (n = 4) were vaccinated i.n. with 103 PFU of WT or LACIV H3N8 CIVs. Mice mock (PBS) vaccinated or vaccinated i.m. with 100 μl of an H3N8 CIV IIV (Nobivac) were used as internal controls. At 10 days p.i., the lungs (A) and spleen (B) were extracted, and the cells were prepared for flow cytometry. Live CD8 T cells specific for NP or PA tetramers were counted. The data represent the means ± the SD of the results for four individual mice. *, WT versus LACIV, Nobivac, or PBS; or *, LACIV versus Nobivac or PBS (P < 0.05 using the Student t test).

We next evaluated the ability of H3N8 LACIV to induce protective immunity. Mice (n = 6) were vaccinated i.n. with 103 PFU of H3N8 WT or LACIV, vaccinated i.m. with 100 μl of the IIV Nobivac, or mock vaccinated with PBS. Two weeks later mice were challenged with 105 PFU of WT H3N8 CIV and viral titers in the lungs of infected mice (n = 3/group) were determined 2 and 4 days after challenge (Fig. 6). Mock-vaccinated mice showed high viral titers in the lungs at days 2 and 4 p.i., whereas mice immunized with H3N8 WT CIV and with LACIV showed no detectable virus in the lungs at those times (Fig. 6). Mice vaccinated with the H3N8 IIV showed high viral titers at day 2 but no detectable virus at day 4 p.i. (Fig. 6).

FIG 6
  • Open in new tab
  • Download powerpoint
FIG 6

Protection efficacy of H3N8 LACIV against homologous viral challenge. Female 5- to-7-week-old C57BL/6 WT mice (n = 6) were vaccinated i.n. with 103 PFU of WT or LACIV H3N8 CIVs. Mice mock (PBS) vaccinated or vaccinated i.m. with 100 μl of an H3N8 CIV IIV (Nobivac) were used as internal controls. At 15 days postvaccination, mice were challenged i.n. with 105 PFU of WT H3N8 CIV. To evaluate viral replication, mice were euthanized at days 2 (n = 3, black) and 4 (n = 3, gray) postchallenge, and the lungs were harvested, homogenized, and used to quantify viral titers by immunofocus assay (FFU/ml). The dotted line indicates the limit of detection (200 FFU/ml). ND, virus not detected. Data represent the mean ± the SD. *, P < 0.05 using the Student t test.

H3N8 LACIV is attenuated in canine tracheal explants compared to H3N8 WT CIV.To compare LACIV and WT H3N8 CIV pathogenicity and replication efficiency at the site of infection within dogs (Fig. 7), we inoculated dog tracheal explants with each virus and compared histological lesions (Fig. 7A), viral NP expression (Fig. 7B), changes in ciliary function (Fig. 7C), and viral replication (Fig. 7D) at different times (days 1, 3, and 5) p.i. The H3N8 WT CIV induced major histological changes in dog tracheal explants, with thinning and desquamation of the epithelium, loss of cilia (Fig. 5A), and significant reduction of ciliary function (Fig. 7C) between days 1 and 5 p.i. Histological damages induced by H3N8 LACIV were delayed and reduced compared to WT H3N8 CIV, since the epithelium maintained its normal thickness until day 3 p.i. (Fig. 7A), and the ciliary function (Fig. 7C) was only significantly reduced from day 3 p.i. Viral kinetics (Fig. 7D) and NP expression (Fig. 7B) were comparable between the two viruses, although only WT H3N8 CIV was detectable at day 1 p.i. (Fig. 7D). Overall, these results indicate that H3N8 LACIV pathogenicity is attenuated in canine tracheal explants compared to its WT counterpart.

FIG 7
  • Open in new tab
  • Download powerpoint
FIG 7

Comparison of WT and LACIV H3N8 CIV infection phenotypes in canine tracheal explants. (A) Histological features of dog tracheal explants infected with 200 PFU of H3N8 WT CIV or H3N8 LACIV or mock-infected with infection media. Lesions are shown in sections stained with H&E. (B) CIV H3N8-infected cells were detected by immunostaining for the viral NP, and positive cells are stained in brown. For both panels A and B, pictures are representatives of three independent experiments, and the black horizontal bars represent 20 μm. (C) Graphical representation of the bead clearance average time of CIV- or mock-infected dog tracheal explants for three independent experiments. Data represent the means ± the SD. ns, P > 0.05 (D1, LACIV versus Mock); *, P < 0.05 (D1, WT versus Mock); **, P < 0.01 (D3, LACIV versus Mock); ***, P < 0.001 (D3, WT versus Mock); ****, P < 0.0001 (D5, LACIV and WT versus Mock). (D) Average viral replication of H3N8 LACIV and H3N8 WT CIV in canine tracheal explants from three independent experiments. Data represent the means ± the SD. The dotted line indicates the limit of detection (20 FFU/ml).

H3N8 LACIV provides limited protection against heterologous H3N2 CIV challenge.We next evaluated whether H3N8 LACIV could induce protective immunity against a heterologous H3N2 CIV challenge (Fig. 8). Mice (n = 6) were vaccinated i.n. with 103 PFU of WT or LACIV H3N8, mock vaccinated with PBS, or vaccinated i.m. with 100 μl of the H3N8 IIV Nobivac or a commercial H3N2 IIV (Zoetis). Antibodies against H3N2 CIV were evaluated by ELISA using cell lysates from mock- or H3N2 CIV-infected MDCK cells as antigens (Fig. 8A). In addition, antibodies against the HA (Fig. 8B) or NA (Fig. 8C) proteins of H3N2 CIV were also evaluated. When the cell lysate was used as an antigen, antibodies against H3N2 CIV were detected in sera of mice vaccinated with WT H3N8 CIV and, to a lower extent, in mice vaccinated with H3N8 LACIV, although the levels were lower than those against H3N8 CIV (Fig. 8A). Similarly, using the recombinant proteins (HA or NA) as antigens to perform the ELISA, only antibodies against H3N2 CIV HA were detected in samples of animals immunized with WT H3N8 CIV (Fig. 8B). However, no antibodies were detected against the NA protein (Fig. 8C). No detectable IgG antibodies against H3N2 CIV were detected in mice vaccinated with the H3N8 IIV Nobivac either using the cell extracts (Fig. 8A) or the recombinant proteins (Fig. 8B and C). The H3N2 CIV IIV induced higher IgG antibodies against H3N2 CIV in all cases, as expected (Fig. 8A to C).

FIG 8
  • Open in new tab
  • Download powerpoint
FIG 8

Immunogenicity and protection efficacy of H3N8 LACIV against heterologous H3N2 CIV challenge. Female 5- to-7-week-old C57BL/6 WT mice were vaccinated i.n. with 103 PFU of WT and LACIV H3N8 CIVs. Mice mock (PBS) vaccinated or vaccinated i.m. with 100 μl of the H3N8 (Nobivac) and an H3N2 CIV IIV (Zoetis) were used as internal controls. (A to C) Antibody cross-reactivity against the heterologous CIV H3N2. At 14 days postvaccination, mice were bled, and sera were evaluated by ELISA for total IgG antibodies against H3N2 CIV proteins using cell extracts of MDCK-infected cells (A). Mock-infected MDCK cell extracts were used to evaluate the specificity of the antibody response. OD, optical density. Data represent the means ± the SD of the results for six individual mice. *, Nobivac versus LACIV; **, WT versus LACIV; or ***, WT versus Nobivac (P < 0.05 using the Student t test). Specific antibody response against recombinant HA (B) and NA (C) proteins from H3N2 CIV were evaluated by ELISA. Data represent the means ± the SD of the results for pooled serum samples. *, WT versus LACIV (P < 0.05 using the Student t test). (D) Protection efficacy of H3N8 LACIV against heterologous H3N2 CIV challenge. At 15 days postvaccination, mice were challenged i.n. with 105 PFU of WT H3N2 CIV. To evaluate WT H3N2 CIV replication, mice were sacrificed at days 2 (n = 3, black) and 4 (n = 3, gray) postchallenge, and the lungs were harvested, homogenized, and used to evaluate the presence of virus by immunofocus assay (FFU/ml). The dotted line indicates the limit of detection (200 FFU/ml). ND, virus not detected. Data represent means ± the SD. *, P < 0.05 using Student t test.

The lower level of cross-reactive immunity against H3N2 CIV upon vaccination with the H3N8 LACIV was confirmed after challenge i.n. with H3N2 CIV 2 weeks postvaccination (Fig. 8D). Mock-vaccinated mice showed high H3N2 CIV titers that were undistinguishable from those seen in animals vaccinated with the H3N8 CIV IIV Nobivac. In contrast, mice vaccinated with the H3N2 CIV IIV showed reduced or undetectable titers at days 2 and 4 postchallenge, respectively. Although we observed similar H3N2 CIV titers at day 2 postchallenge, viral titers at day 4 p.i. in mice vaccinated with the H3N8 LACIV were ∼100 times lower than those obtained in the mock-vaccinated group. These results suggest that H3N8 LACIV can induce limited cross-reactive immune responses and heterologous protection, most probably mediated by a T cell response, against H3N2 CIV but that the efficacy is lower than that obtained with the H3N2 IIV.

DISCUSSION

We report here a novel LAIV prepared using plasmid-based reverse genetics techniques, which may be used for the prevention of H3N8 CIV. We generated a recombinant H3N8 CIV containing the mutations responsible for the ts phenotype of the human A/AA/6/60 H2N2 LAIV, resulting in ts H3N8 CIV (LACIV) that was highly attenuated in vivo and ex vivo compared to its WT counterpart. Our H3N8 LACIV was able to confer, upon a single i.n. immunization in mice, complete protection against challenge with WT H3N8 CIV. This demonstrates the feasibility of using the ts H3N8 LACIV as a safe, immunogenic, and protective vaccine to control H3N8 CIV in dogs. However, the H3N8 LACIV showed limited immunogenicity and protection efficacy against the heterologous H3N2 CIV, suggesting the need of a different LACIV for the treatment and control of H3N2 CIV.

The ts, ca, and att A/AA/6/60 H2N2 LAIV has been licensed for human use since 2003, and is used as a master donor virus for the generation of both seasonal or potentially pandemic human LAIV by creating reassortant viruses containing the six H2N2-derived internal viral RNA (vRNA) segments (PB2, PB1, PA, NP, M, and NS) and the two glycoprotein-encoding vRNAs (HA and NA) from a virus that antigenically matches the strains predicted to circulate in the upcoming influenza season (seasonal vaccine) or potentially pandemic strains (pandemic vaccine) (44, 45). Five mutations (PB2 N265S; PB1 K391E, D581G, and A661T; and NP D34G) are responsible for the ts phenotype of the A/AA/6/60 H2N2 LAIV, and those mutations also impart a strong ts phenotype and attenuation to other viral strains, such as PR8 (36, 46) and pH1N1 (37).

Intranasal immunization is a desirable delivery method for providing optimal immunity to IAV because it leads to the generation of a mucosal immune responses, creating an immune barrier at the site of potential infection (47), as well as systemic humoral responses, cellular immunity (45, 48–51). Similar to infection with WT IAV, LAIV immunization also leads to recruitment of influenza-specific CD8 T cells to the lungs (42, 50, 52–54), which provides immunity against heterologous influenza challenge (42, 52). Thus, a LAIV rather than an IIV is desired for the control of IAV infections (22, 23, 27, 55).

Since the emergence of H3N8 CIV in 1999 in the United States and the H3N2 CIV in Asia (in 2005) and the United States (in 2015), CIVs have been maintained mainly in animal shelters and kennels as those populations allow ready transmission of the virus (10, 16, 17, 56, 57). Strains of the H3 subtype of IAV infect a number of mammalian hosts, including humans, pigs, horses, dogs, cats, and seals, as well as poultry (6, 11, 57–61). Naturally occurring H3N1 virus carrying the HA gene of an avian-like H3N2 CIV and the other seven segments of the human pH1N1 has been reported in dogs in Korea (20), suggesting that dogs could act as an intermediate host for genetic reassortment of IAV, including those that might infect humans. However, no transmission of H3N8 or H3N2 CIV transmission from dogs to humans has been reported to date. It may be possible to eradicate both H3N8 and H3N2 CIVs from the dog population through infection control, as well as by using vaccination approaches. CIV LAIVs represent a better option for efficient CIV control and probably eradication since they induce better and faster antiviral immunity.

The H3N8 LACIV generated here was ts and attenuated in mice and induced protective immune responses against challenge with homologous H3N8 CIV WT, and the responses were stronger than those obtained with a commercial H3N8 CIV IIV. Its replication and pathogenesis were also restricted in canine tracheal explants, and we are currently evaluating the safety, immunogenicity, and protection efficacy of our H3N8 LACIV in dogs, the real target population.

To achieve protection in dogs, animals are vaccinated i.m. with 1 ml of the CIV H3N8 IIV (Nobivac) using a two-dose regime (62). The average weight of a male or female mouse is ∼20 g, whereas the average weight of a female or male beagle dog is ∼10 kg. Thus, in principle mice should be vaccinated with 500 times less (just 2 μl) of the CIV H3N8 IIV than dogs. However, in our experiments, mice were immunized i.m. with 100 μl of the CIV H3N8 IIV, a 50× higher dose than on a weight basis. In addition, to evaluate the amount of antigen in the CIV H3N8 IIV, we performed an hemagglutination assay using the commercial CIV H3N8 IIV or our CIV H3N8 LAIV. The assay showed that mice vaccinated with the CIV H3N8 IIV were inoculated with approximately 106 viral particles/mouse of inactivated virus, 1,000 times more than the dose (i.e., 103) of the H3N8 CIV LAIV. The LAIV still elicited better antibody responses and protection compared to the CIV H3N8 IIV.

Segment eight of IAV encodes the NS1 viral protein, which controls the adaptive immune responses by inhibiting the interferon-antiviral response of the host (63). Therefore, NS1 is a virulence factor that offers an attractive target for the development of attenuated viruses as LAIVs. In fact, IAVs harboring a truncated-NS1 have been shown as promising vaccines candidates (28, 64–71). In a recent work, we have generated recombinant H3N8 CIVs containing truncated (NS1-126, NS1-99, or NS1-73) or deleted (ΔNS1) NS1 proteins and tested them as potential LAIVs against CIV H3N8 infections (28). The recombinant NS1 mutant H3N8 CIVs were attenuated in vivo (mice) and in vitro (dog tracheal explants) but were able to confer complete protection against challenge with WT CIV H3N8 (28). Moreover, the immunogenicity and protection efficacy of our NS1 mutant H3N8 CIVs was also better than that observed with an H3N8 CIV IIV (28). Future research should determine which one of these attenuation strategies (NS1 mutant or ts H3N8 CIVs) is more efficient for their implementation as a LAIV for the prevention and control of H3N8 CIV in dogs.

MATERIALS AND METHODS

Cells and viruses.Human embryonic kidney 293T cells (293T; ATCC CRL-11268) and Madin-Darby canine kidney cells (MDCK; ATCC CCL-34) were grown at 37°C with 5% CO2 in Dulbecco modified Eagle medium (DMEM; Mediatech, Inc.) supplemented with 10% fetal bovine serum (FBS) and 1% PSG (penicillin, 100 U/ml; streptomycin 100 μg/ml; l-glutamine, 2 mM) (40).

Recombinant wild-type (WT) and live-attenuated (LACIV) H3N8 CIVs were generated using A/canine/NY/dog23/2009 H3N8 plasmid-based reverse genetics techniques (72) and grown in MDCK cells at 33°C. Influenza A/Ca/IL/41915/2015 H3N2 was also grown in MDCK cells at 33°C. For infections, virus stocks were diluted in PBS, 0.3% bovine albumin BA, and 1% PS (PBS/BA/PS). After viral infections, cells were maintained in DMEM with 0.3% BA, 1% PSG, and 1 μg/ml TPCK (tolylsulfonyl phenylalanyl chloromethyl ketone)-treated trypsin (Sigma) (38).

Plasmids.To generate H3N8 LACIV, the PB2 and PB1 genes were subcloned in a pUC19 plasmid (New England BioLabs), and then ts mutations (PB2 N265S and PB1 K391E, D581G, and A661T) were introduced by site-directed mutagenesis. The presence of introduced mutations was confirmed by sequencing. Mutated PB2 and PB1 viral segments were subcloned from pUC19 into the ambisense pDZ plasmid for virus rescue. To test the ability of WT and LACIV H3N8 polymerases to replicate and transcribe at different temperatures (33, 37, and 39°C) using a minigenome assay, we engineered a pPolI plasmid containing the canine RNA polymerase I (Pol-I) promoter and the mouse Pol-I terminator separated by SapI endonuclease restriction sites (cpPol-I). The canine Pol-I promoter was obtained by PCR from MDCK cells (73). Then, the Gaussia luciferase (Gluc) reporter gene containing the 3′ and the 5′ noncoding regions of the viral NP (v)RNA was cloned into the cpPol-I plasmid to generate the cpPol-I Gluc reporter plasmid. All plasmids were confirmed by sequencing (ACGT Inc.). Primers for the generation of the different plasmids are available upon request.

Minigenome assays.MDCK cells (12-well plate format, 5 × 105 cells/well, triplicates) were cotransfected in suspension using Lipofectamine 2000 with 250 ng of each of the H3N8 WT or LACIV ambisense pDZ PB2, PB1, PA, and NP plasmids, together with 500 ng of the cpPol-I Gluc plasmid. A mammalian expression pCAGGS plasmid encoding Cypridina luciferase (Cluc; 100 ng) was also included to normalize transfection efficiencies (74). Cells transfected in the absence of the pDZ NP plasmid were used as negative control. At 24 h posttransfection, the Gluc and Cluc expression levels were determined using a luciferase assay kit (New England BioLabs) and quantified with a Lumicount luminometer (Packard). The fold induction over the level of induction for the negative control (the absence of NP) was determined. The mean values and standard deviations (SD) were calculated and statistical analysis was performed using a two-tailed Student t test using Microsoft Excel software.

Virus rescue.Virus rescues were performed as previously described (40, 75). Briefly, cocultures (1:1) of 293T/MDCK cells (6-well plate format, 106 cells/well, triplicates) were cotransfected in suspension, using Lipofectamine 2000 (Invitrogen), with 1 μg of the eight-ambisense H3N8 WT CIV (pDZ-PB2, -PB1, -PA, -HA, -NP, -NA, -M, and –NS) plasmids. To rescue the H3N8 LACIV, WT PB2 and PB1 pDZ plasmids were substituted by those containing PB2 and PB1 H3N8 LACIV. At 12 h posttransfection, the transfection medium was replaced with postinfection (p.i.) medium containing DMEM supplemented with 0.3% BSA, 1% PSG, and 0.5 μg/ml TPCK-treated trypsin (Sigma). Tissue culture supernatants (TCS) were collected 3 days posttransfection, clarified, and used to infect fresh monolayers of MDCK cells (6-well plate format, 106 cells/well, triplicates). At 3 days p.i., recombinant viruses were plaque purified and scaled up using MDCK cells at 33°C (40). Virus stocks were titrated by standard plaque assay (PFU/ml) in MDCK cells at 33°C (40).

Virus growth kinetics.Multicycle growth analyses were performed by infecting confluent monolayers of MDCK cells (12-well plate format, 5 × 105 cells/well, triplicates) at an MOI of 0.001. Viral titers in TCS collected at various times p.i. were determined by immunofocus assay (fluorescent forming units [FFU]/ml) in MDCK cells as previously described (40). Briefly, confluent MDCK cells (96-well plate format, 5 ×104 cells/well, triplicates) were infected with 10-fold serial dilutions of H3N8 WT or LACIV. At 12 h p.i., the cells were fixed and permeabilized (4% formaldehyde and 0.5% Triton X-100 in PBS) for 15 min at room temperature. After being washed with PBS, the cells were incubated in blocking solution (2.5% BSA in PBS) for 1 h at room temperature and then incubated with 1 μg/ml of an anti-NP monoclonal antibody (HB-65; ATTC) for 1 h at 37°C. After a washing step with PBS, the cells were incubated with FITC-conjugated secondary anti-mouse antibody (Dako) for 1 h at 37°C. The mean values and SD were calculated using Microsoft Excel software.

Animal experiments.Adult (5- to 7-week-old) female WT C57BL/6 mice were purchased from the National Cancer Institute (NCI) and maintained in the animal care facility at the University of Rochester under specific-pathogen-free conditions. Animal 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 (76). Mice were anesthetized intraperitoneally with 2,2,2-tribromoethanol (Avertin; 240 mg/kg [body weight]) and then inoculated i.n. with 30 μl of the indicated amounts of H3N8 WT or LACIV or H3N2 WT. Alternatively, 100 μl of a commercially available inactivated H3N8 CIV vaccine (Nobivac; Merck Animal Health) or inactivated H3N2 CIV vaccine (Zoetis) were inoculated i.m. Virus replication was determined by measuring viral titers in the lungs of infected mice at the indicated days p.i. To that end, three mice in each group were euthanized by administration of a lethal dose of Avertin and exsanguination, and the lungs were collected and homogenized. Virus titers were determined by immunofocus assay (FFU/ml) as indicated above. Mouse sera were collected by submandibular bleeding 24 h prior to viral challenges and evaluated for the presence of influenza virus antibodies by ELISAs and neutralizing antibodies by HAI assays.

Evaluation of T cells response in lung and spleen.Mice (n = 4) were immunized as described above: 103 PFU of H3N8 WT or LACIV, mock vaccinated with PBS, or vaccinated with 100 μl of Nobivac (IIV).

Cellular preparations.At 10 days p.i., the lungs and spleens were perfused with PBS, removed, and separated into right and left lobes. Lung tissue was dissociated in C tubes by the GentleMACS (Miltenyi Biotek) using the Lung01 program. Samples were incubated in 5 ml (2 μg/ml) of Collagenase II in RPMI plus 8% FBS at 37°C for 30 min, with gentle agitation every 10 min. After digestion, samples were further dissociated using the Heart01 program. Cell suspensions were filtered through 70-μm-pore size filters prior to 75:40 Percoll (GE Healthcare) discontinuous gradient separation. The top layer, containing fat and other debris, was removed by aspiration. The cell layer was harvested and washed prior to counting and staining. Single-cell suspensions were prepared from collected spleens by disruption in RPMI plus 8% FBS. Counting was achieved through trypan blue exclusion on a hemocytometer.

Flow cytometry.Single cell suspensions were stained in PBS containing 1% FBS, purified CD16/32 (clone 2.4G2), NP and PA tetramers (43), and the following antibodies: TCRβ-PerCPCy5.5, CD8α-FITC, CD4-BV421, CD44-APCCy7, and CD62L-PECy7. Cells were subsequently stained for viability using Live/Dead Aqua (Invitrogen). All antibodies were obtained from eBioscience, BD Biosciences, or BioLegend. PA and NP tetramers were obtained from the NIH tetramer core facility (Atlanta, GA). Cells were analyzed by an LSRII (BD Biosciences) in the University of Rochester Flow Cytometry core facility and analyzed using FlowJo software (Tree Star).

ELISAs.ELISAs were performed as previously described (40) by coating 96-well plates at 4°C for 16 h with lysates from mock- or H3N8 or H3N2 WT CIV-infected MDCK cells or with H3N2 CIV HA (250 ng per well [IRR catalog no. FR-1478]) or NA (250 ng per well [IRR catalog no. FR-1479]). After blocking with 1% BSA for 1 h at room temperature, the plates were incubated with 2-fold serial dilutions (starting dilution of 1:50) of mouse sera for 1 h at 37°C. After incubation, the plates were washed with H2O and incubated with a horseradish peroxidase-conjugated goat anti-mouse IgG (1:2,000; Southern Biotech) for 1 h at 37°C. Reactions were then developed with tetramethylbenzidine (TMB) substrate (BioLegend) for 10 min at room temperature, quenched with 2N H2SO4, and read at 450 nm (Vmax kinetic microplate reader; Molecular Devices).

HAI assays.To evaluate the presence of H3N8 CIV neutralizing antibodies, mous sera were treated with receptor-destroying enzyme (RDE; Denka Seiken) and heat inactivated for 30 min at 56°C. The sera were then serially 2-fold diluted (starting dilution of 1:50) in 96-well V-bottom plates and mixed 1:1 with 4 hemagglutinating units (HAU) of WT H3N8 CIV for 30 min at room temperature. The HAI titers were determined by adding 0.5% turkey red blood cells to the virus-antibody mixtures for 30 min on ice, as previously described (40). The geometric mean titers and SD from individual mice (n = 6) were calculated from the last well where hemagglutination was inhibited.

Canine tracheal explants preparation and virus titrations.Three dog tracheas were harvested from healthy beagles (Charles River Laboratories) that had been used as negative controls in unrelated studies. Briefly, tracheas were collected aseptically immediately upon euthanasia and transported in prewarmed medium as previously described (18). Tracheas were washed five times for a total period of 4 h and maintained at 33°C, 5% CO2, and 95% humidity between washes. The connective tissue was removed, and the trachea was then open lengthwise. Each tracheal ring was divided in four 0.25-cm2 explants and transferred with the epithelium facing upward onto an agarose plug covered by a sterile filter. The explants were kept for a total of 6 days at 33°C, 5% CO2, and 95% humidity.

Tracheal explants were infected after a period of 24 h postpreparation (designed as day zero) with 200 PFU of WT or LACIV H3N8 or mock infected with culture medium. Inoculated explants were sampled for virus quantification, bead clearance assays, and histology at days 0, 1, 3, and 5 p.i. Viral replication was evaluated by plaque assays on MDCK cells.

Estimation of bead clearance time.The ciliary function of tracheal explants was evaluated as previously described (18) by placing 5 μl of polystyrene microsphere beads (Polysciences, Northampton, United Kingdom) on the explant apical surfaces and measuring the time to displace the beads.

Histological analysis and immunohistochemistry.After collection, the explants were fixed in 10% buffered formalin for a minimum of 48 h before paraffin embedding and sectioning. Subsequently, 4-μm sections of paraffin embedded tissue were either stained with hematoxylin and eosin (H&E) for histopathological evaluation or immunostained for the viral NP using standard procedures as previously described (18). For NP immunostaining, the Dako supervision system was used according to the manufacturer's instructions, along with a monoclonal mouse anti-NP (clone HB65; dilution, 1:500). Slides were counterstained with Mayer's hematoxylin. Histological images were captured with cellD software (Olympus).

ACKNOWLEDGMENTS

This research was partially funded by the University of Rochester Technology Development Fund to L.M.-S. and C.R.P. C.C. was supported by the Horserace Betting Levy Board, and P.R.M. was supported by the Medical Research Council of the United Kingdom (grant MC_UU_120/14/9).

FOOTNOTES

    • Received 8 November 2016.
    • Accepted 5 December 2016.
    • Accepted manuscript posted online 7 December 2016.
  • Copyright © 2017 American Society for Microbiology.

All Rights Reserved .

REFERENCES

  1. 1.↵
    1. Baker SF,
    2. Nogales A,
    3. Martinez-Sobrido L
    . 2015. Downregulating viral gene expression: codon usage bias manipulation for the generation of novel influenza A virus vaccines. Future Virol10:715–730. doi:10.2217/fvl.15.31.
    OpenUrlCrossRef
  2. 2.↵
    1. Varghese JN,
    2. McKimm-Breschkin JL,
    3. Caldwell JB,
    4. Kortt AA,
    5. Colman PM
    . 1992. The structure of the complex between influenza virus neuraminidase and sialic acid, the viral receptor. Proteins14:327–332. doi:10.1002/prot.340140302.
    OpenUrlCrossRefPubMedWeb of Science
  3. 3.↵
    1. Carrat F,
    2. Flahault A
    . 2007. Influenza vaccine: the challenge of antigenic drift. Vaccine25:6852–6862. doi:10.1016/j.vaccine.2007.07.027.
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    1. Palese PS,
    2. ML
    . 2007. Orthomyxoviridae: the viruses and their replication, p 1647–1690. InKnipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA (ed), Fields virology, 5th ed. Lippincott/Williams & Wilkins, Philadelphia, PA.
  5. 5.↵
    1. Tong S,
    2. Zhu X,
    3. Li Y,
    4. Shi M,
    5. Zhang J,
    6. Bourgeois M,
    7. Yang H,
    8. Chen X,
    9. Recuenco S,
    10. Gomez J,
    11. Chen LM,
    12. Johnson A,
    13. Tao Y,
    14. Dreyfus C,
    15. Yu W,
    16. McBride R,
    17. Carney PJ,
    18. Gilbert AT,
    19. Chang J,
    20. Guo Z,
    21. Davis CT,
    22. Paulson JC,
    23. Stevens J,
    24. Rupprecht CE,
    25. Holmes EC,
    26. Wilson IA,
    27. Donis RO
    . 2013. New world bats harbor diverse influenza A viruses. PLoS Pathog9:e1003657. doi:10.1371/journal.ppat.1003657.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. de Jong JC,
    2. Smith DJ,
    3. Lapedes AS,
    4. Donatelli I,
    5. Campitelli L,
    6. Barigazzi G,
    7. Van Reeth K,
    8. Jones TC,
    9. Rimmelzwaan GF,
    10. Osterhaus AD,
    11. Fouchier RA
    . 2007. Antigenic and genetic evolution of swine influenza A (H3N2) viruses in Europe. J Virol81:4315–4322. doi:10.1128/JVI.02458-06.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Taubenberger JK,
    2. Kash JC
    . 2010. Influenza virus evolution, host adaptation, and pandemic formation. Cell Host Microbe7:440–451. doi:10.1016/j.chom.2010.05.009.
    OpenUrlCrossRefPubMedWeb of Science
  8. 8.↵
    1. Webster RG,
    2. Bean WJ,
    3. Gorman OT,
    4. Chambers TM,
    5. Kawaoka Y
    . 1992. Evolution and ecology of influenza A viruses. Microbiol Rev56:152–179.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Yoon SW,
    2. Webby RJ,
    3. Webster RG
    . 2014. Evolution and ecology of influenza a viruses. Curr Top Microbiol Immunol385:359–375.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Crawford PC,
    2. Dubovi EJ,
    3. Castleman WL,
    4. Stephenson I,
    5. Gibbs EP,
    6. Chen L,
    7. Smith C,
    8. Hill RC,
    9. Ferro P,
    10. Pompey J,
    11. Bright RA,
    12. Medina MJ,
    13. Johnson CM,
    14. Olsen CW,
    15. Cox NJ,
    16. Klimov AI,
    17. Katz JM,
    18. Donis RO
    . 2005. Transmission of equine influenza virus to dogs. Science310:482–485. doi:10.1126/science.1117950.
    OpenUrlAbstract/FREE Full Text
  11. 11.↵
    1. Song D,
    2. Kang B,
    3. Lee C,
    4. Jung K,
    5. Ha G,
    6. Kang D,
    7. Park S,
    8. Park B,
    9. Oh J
    . 2008. Transmission of avian influenza virus (H3N2) to dogs. Emerg Infect Dis14:741–746. doi:10.3201/eid1405.071471.
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    JAVMA News. 2015. Outbreak of canine influenza caused by new strain of virus. J Am Vet Med Assoc246:1049–1049.
    OpenUrl
  13. 13.↵
    1. Jeoung HY,
    2. Lim SI,
    3. Shin BH,
    4. Lim JA,
    5. Song JY,
    6. Song DS,
    7. Kang BK,
    8. Moon HJ,
    9. An DJ
    . 2013. A novel canine influenza H3N2 virus isolated from cats in an animal shelter. Vet Microbiol165:281–286. doi:10.1016/j.vetmic.2013.03.021.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Song DS,
    2. An DJ,
    3. Moon HJ,
    4. Yeom MJ,
    5. Jeong HY,
    6. Jeong WS,
    7. Park SJ,
    8. Kim HK,
    9. Han SY,
    10. Oh J S,
    11. Park BK,
    12. Kim JK,
    13. Poo H,
    14. Webster RG,
    15. Jung K,
    16. Kang BK
    . 2011. Interspecies transmission of the canine influenza H3N2 virus to domestic cats in South Korea. J Gen Virol92:2350–2355. doi:10.1099/vir.0.033522-0.
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    1. Yoon KJ,
    2. Cooper VL,
    3. Schwartz KJ,
    4. Harmon KM,
    5. Kim WI,
    6. Janke BH,
    7. Strohbehn J,
    8. Butts D,
    9. Troutman J
    . 2005. Influenza virus infection in racing greyhounds. Emerg Infect Dis11:1974–1976. doi:10.3201/eid1112.050810.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    1. Holt DE,
    2. Mover MR,
    3. Brown DC
    . 2010. Serologic prevalence of antibodies against canine influenza virus (H3N8) in dogs in a metropolitan animal shelter. J Am Vet Med Assoc237:71–73. doi:10.2460/javma.237.1.71.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Pecoraro HL,
    2. Spindel ME,
    3. Bennett S,
    4. Lunn KF,
    5. Landolt GA
    . 2013. Evaluation of virus isolation, one-step real-time reverse transcription polymerase chain reaction assay, and two rapid influenza diagnostic tests for detecting canine Influenza A virus H3N8 shedding in dogs. J Vet Diagn Invest25:402–406. doi:10.1177/1040638713480500.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Gonzalez G,
    2. Marshall JF,
    3. Morrell J,
    4. Robb D,
    5. McCauley JW,
    6. Perez DR,
    7. Parrish CR,
    8. Murcia PR
    . 2014. Infection and pathogenesis of canine, equine, and human influenza viruses in canine tracheas. J Virol88:9208–9219. doi:10.1128/JVI.00887-14.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. Song D,
    2. Kim H,
    3. Na W,
    4. Hong M,
    5. Park SJ,
    6. Moon H,
    7. Kang B,
    8. Lyoo KS,
    9. Yeom M,
    10. Jeong DG,
    11. An DJ,
    12. Kim JK
    . 2015. Canine susceptibility to human influenza viruses (A/pdm 09H1N1, A/H3N2 and B). J Gen Virol96:254–258. doi:10.1099/vir.0.070821-0.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Song D,
    2. Moon HJ,
    3. An DJ,
    4. Jeoung HY,
    5. Kim H,
    6. Yeom MJ,
    7. Hong M,
    8. Nam JH,
    9. Park SJ,
    10. Park BK,
    11. Oh J S,
    12. Song M,
    13. Webster RG,
    14. Kim JK,
    15. Kang BK
    . 2012. A novel reassortant canine H3N1 influenza virus between pandemic H1N1 and canine H3N2 influenza viruses in Korea. J Gen Virol93:551–554. doi:10.1099/vir.0.037739-0.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Yen HL,
    2. Webster RG
    . 2009. Pandemic influenza as a current threat. Curr Top Microbiol Immunol333:3–24.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Pica N,
    2. Palese P
    . 2013. Toward a universal influenza virus vaccine: prospects and challenges. Annu Rev Med64:189–202. doi:10.1146/annurev-med-120611-145115.
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    1. Wong SS,
    2. Webby RJ
    . 2013. Traditional and new influenza vaccines. Clin Microbiol Rev26:476–492. doi:10.1128/CMR.00097-12.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Belshe RB,
    2. Edwards KM,
    3. Vesikari T,
    4. Black SV,
    5. Walker RE,
    6. Hultquist M,
    7. Kemble G,
    8. Connor EM,
    9. CAIV-T Comparative Efficacy Study Group
    . 2007. Live attenuated versus inactivated influenza vaccine in infants and young children. N Engl J Med356:685–696. doi:10.1056/NEJMoa065368.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    1. Cox MM,
    2. Patriarca PA,
    3. Treanor J
    . 2008. FluBlok, a recombinant hemagglutinin influenza vaccine. Influenza Other Respir Viruses2:211–219. doi:10.1111/j.1750-2659.2008.00053.x.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Osterholm MT,
    2. Kelley NS,
    3. Sommer A,
    4. Belongia EA
    . 2012. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis12:36–44. doi:10.1016/S1473-3099(11)70295-X.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    1. Pronker ES,
    2. Claassen E,
    3. Osterhaus AD
    . 2012. Development of new generation influenza vaccines: recipes for success?Vaccine30:7344–7347. doi:10.1016/j.vaccine.2012.09.071.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Nogales A,
    2. Huang K,
    3. Chauche C,
    4. DeDiego ML,
    5. Murcia PR,
    6. Parrish CR,
    7. Martinez-Sobrido L
    . 2017. Canine influenza viruses with modified NS1 proteins for the development of live-attenuated vaccines. Virology500:1–10. doi:10.1016/j.virol.2016.10.008.
    OpenUrlCrossRef
  29. 29.↵
    1. Belongia EA,
    2. Kieke BA,
    3. Donahue JG,
    4. Greenlee RT,
    5. Balish A,
    6. Foust A,
    7. Lindstrom S,
    8. Shay DK
    . 2009. Effectiveness of inactivated influenza vaccines varied substantially with antigenic match from the 2004-2005 season to the 2006-2007 season. J Infect Dis199:159–167. doi:10.1086/595861.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    1. Gorse GJ,
    2. Belshe RB,
    3. Munn NJ
    . 1991. Superiority of live attenuated compared with inactivated influenza A virus vaccines in older, chronically ill adults. Chest100:977–984. doi:10.1378/chest.100.4.977.
    OpenUrlCrossRefPubMedWeb of Science
  31. 31.↵
    1. Maassab HF
    . 1968. Plaque formation of influenza virus at 25°C. Nature219:645–646.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Chan W,
    2. Zhou H,
    3. Kemble G,
    4. Jin H
    . 2008. The cold adapted and temperature sensitive influenza A/Ann Arbor/6/60 virus, the master donor virus for live attenuated influenza vaccines, has multiple defects in replication at the restrictive temperature. Virology380:304–311. doi:10.1016/j.virol.2008.07.027.
    OpenUrlCrossRefPubMedWeb of Science
  33. 33.↵
    1. Cox NJ,
    2. Kitame F,
    3. Kendal AP,
    4. Maassab HF,
    5. Naeve C
    . 1988. Identification of sequence changes in the cold-adapted, live attenuated influenza vaccine strain, A/Ann Arbor/6/60 (H2N2). Virology167:554–567. doi:10.1016/S0042-6822(88)90118-3.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Snyder MH,
    2. Betts RF,
    3. DeBorde D,
    4. Tierney EL,
    5. Clements ML,
    6. Herrington D,
    7. Sears SD,
    8. Dolin R,
    9. Maassab HF,
    10. Murphy BR
    . 1988. Four viral genes independently contribute to attenuation of live influenza A/Ann Arbor/6/60 (H2N2) cold-adapted reassortant virus vaccines. J Virol62:488–495.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Cox A,
    2. Baker SF,
    3. Nogales A,
    4. Martinez-Sobrido L,
    5. Dewhurst S
    . 2015. Development of a mouse-adapted live attenuated influenza virus that permits in vivo analysis of enhancements to the safety of live attenuated influenza virus vaccine. J Virol89:3421–3426. doi:10.1128/JVI.02636-14.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. Jin H,
    2. Zhou H,
    3. Lu B,
    4. Kemble G
    . 2004. Imparting temperature sensitivity and attenuation in ferrets to A/Puerto Rico/8/34 influenza virus by transferring the genetic signature for temperature sensitivity from cold-adapted A/Ann Arbor/6/60. J Virol78:995–998. doi:10.1128/JVI.78.2.995-998.2004.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Zhou B,
    2. Li Y,
    3. Speer SD,
    4. Subba A,
    5. Lin X,
    6. Wentworth DE
    . 2012. Engineering temperature-sensitive live attenuated influenza vaccines from emerging viruses. Vaccine30:3691–3702. doi:10.1016/j.vaccine.2012.03.025.
    OpenUrlCrossRefPubMed
  38. 38.↵
    1. Martinez-Sobrido L,
    2. Garcia-Sastre A
    . 2010. Generation of recombinant influenza virus from plasmid DNA. J Vis Exp2010:2057. doi:10.3791/2057.
    OpenUrlCrossRef
  39. 39.↵
    1. Jin H,
    2. Lu B,
    3. Zhou H,
    4. Ma C,
    5. Zhao J,
    6. Yang CF,
    7. Kemble G,
    8. 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. Virology306:18–24. doi:10.1016/S0042-6822(02)00035-1.
    OpenUrlCrossRefPubMedWeb of Science
  40. 40.↵
    1. Nogales A,
    2. Baker SF,
    3. Ortiz-Riano E,
    4. Dewhurst S,
    5. Topham DJ,
    6. Martinez-Sobrido L
    . 2014. Influenza A virus attenuation by codon deoptimization of the NS gene for vaccine development. J Virol88:10525–10540. doi:10.1128/JVI.01565-14.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    1. Nogales A,
    2. DeDiego ML,
    3. Topham DJ,
    4. Martinez-Sobrido L
    . 2016. Rearrangement of influenza virus spliced segments for the development of live-attenuated vaccines. J Virol90:6291–6302. doi:10.1128/JVI.00410-16.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Baker SF,
    2. Guo H,
    3. Albrecht RA,
    4. Garcia-Sastre A,
    5. Topham DJ,
    6. Martinez-Sobrido L
    . 2013. Protection against lethal influenza with a viral mimic. J Virol87:8591–8605. doi:10.1128/JVI.01081-13.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    1. Reilly EC,
    2. Lambert-Emo K,
    3. Topham DJ
    . 2016. The effects of acute neutrophil depletion on resolution of acute influenza infection, establishment of tissue resident memory (TRM), and heterosubtypic immunity. PLoS One11:e0164247. doi:10.1371/journal.pone.0164247.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Maassab HF,
    2. Bryant ML
    . 1999. The development of live attenuated cold-adapted influenza virus vaccine for humans. Rev Med Virol9:237–244. doi:10.1002/(SICI)1099-1654(199910/12)9:4<237::AID-RMV252>3.0.CO;2-G.
    OpenUrlCrossRefPubMedWeb of Science
  45. 45.↵
    1. Murphy BR,
    2. Coelingh K
    . 2002. Principles underlying the development and use of live attenuated cold-adapted influenza A and B virus vaccines. Viral Immunol15:295–323. doi:10.1089/08828240260066242.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Cox A,
    2. Baker SF,
    3. Nogales A,
    4. Martinez-Sobrido L,
    5. Dewhurst S
    . 2014. Development of a mouse-adapted, live-attenuated influenza virus that permits in vivo analysis of enhancements to the safety of LAIV. J Virol89:3421–3426. doi:10.1128/JVI.02636-14.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Kohlmeier JE,
    2. Woodland DL
    . 2009. Immunity to respiratory viruses. Annu Rev Immunol27:61–82. doi:10.1146/annurev.immunol.021908.132625.
    OpenUrlCrossRefPubMedWeb of Science
  48. 48.↵
    1. Cheng X,
    2. Zengel JR,
    3. Suguitan AL Jr,
    4. Xu Q,
    5. Wang W,
    6. Lin J,
    7. Jin H
    . 2013. Evaluation of the humoral and cellular immune responses elicited by the live attenuated and inactivated influenza vaccines and their roles in heterologous protection in ferrets. J Infect Dis208:594–602. doi:10.1093/infdis/jit207.
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. De Villiers PJ,
    2. Steele AD,
    3. Hiemstra LA,
    4. Rappaport R,
    5. Dunning AJ,
    6. Gruber WC,
    7. Forrest BD
    . 2009. Efficacy and safety of a live attenuated influenza vaccine in adults 60 years of age and older. Vaccine28:228–234. doi:10.1016/j.vaccine.2009.09.092.
    OpenUrlCrossRefPubMedWeb of Science
  50. 50.↵
    1. Katsura H,
    2. Iwatsuki-Horimoto K,
    3. Fukuyama S,
    4. Watanabe S,
    5. Sakabe S,
    6. Hatta Y,
    7. Murakami S,
    8. Shimojima M,
    9. Horimoto T,
    10. Kawaoka Y
    . 2012. A replication-incompetent virus possessing an uncleavable hemagglutinin as an influenza vaccine. Vaccine30:6027–6033. doi:10.1016/j.vaccine.2012.07.059.
    OpenUrlCrossRefPubMed
  51. 51.↵
    1. Victor ST,
    2. Watanabe S,
    3. Katsura H,
    4. Ozawa M,
    5. Kawaoka Y
    . 2012. A replication-incompetent PB2-knockout influenza A virus vaccine vector. J Virol89:3421–3426. doi:10.1128/JVI.02636-14.
    OpenUrlCrossRef
  52. 52.↵
    1. Guo H,
    2. Baker SF,
    3. Martinez-Sobrido L,
    4. Topham DJ
    . 2014. Induction of CD8 T cell heterologous protection by a single dose of single-cycle infectious influenza virus. J Virol88:12006–12016. doi:10.1128/JVI.01847-14.
    OpenUrlAbstract/FREE Full Text
  53. 53.↵
    1. Powell TJ,
    2. Silk JD,
    3. Sharps J,
    4. Fodor E,
    5. Townsend AR
    . 2012. Pseudotyped influenza A virus as a vaccine for the induction of heterotypic immunity. J Virol86:13397–13406. doi:10.1128/JVI.01820-12.
    OpenUrlAbstract/FREE Full Text
  54. 54.↵
    1. Uraki R,
    2. Kiso M,
    3. Iwatsuki-Horimoto K,
    4. Fukuyama S,
    5. Takashita E,
    6. Ozawa M,
    7. Kawaoka Y
    . 2013. A novel bivalent vaccine based on a PB2-knockout influenza virus protects mice from pandemic H1N1 and highly pathogenic H5N1 virus challenges. J Virol87:7874–7881. doi:10.1128/JVI.00076-13.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    1. Hoft DF,
    2. Babusis E,
    3. Worku S,
    4. Spencer CT,
    5. Lottenbach K,
    6. Truscott SM,
    7. Abate G,
    8. Sakala IG,
    9. Edwards KM,
    10. Creech CB,
    11. Gerber MA,
    12. Bernstein DI,
    13. Newman F,
    14. Graham I,
    15. Anderson EL,
    16. Belshe RB
    . 2011. Live and inactivated influenza vaccines induce similar humoral responses, but only live vaccines induce diverse T-cell responses in young children. J Infect Dis204:845–853. doi:10.1093/infdis/jir436.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Hayward JJ,
    2. Dubovi EJ,
    3. Scarlett JM,
    4. Janeczko S,
    5. Holmes EC,
    6. Parrish CR
    . 2010. Microevolution of canine influenza virus in shelters and its molecular epidemiology in the United States. J Virol84:12636–12645. doi:10.1128/JVI.01350-10.
    OpenUrlAbstract/FREE Full Text
  57. 57.↵
    1. Rivailler P,
    2. Perry IA,
    3. Jang Y,
    4. Davis CT,
    5. Chen LM,
    6. Dubovi EJ,
    7. Donis RO
    . 2010. Evolution of canine and equine influenza (H3N8) viruses cocirculating between 2005 and 2008. Virology408:71–79. doi:10.1016/j.virol.2010.08.022.
    OpenUrlCrossRefPubMedWeb of Science
  58. 58.↵
    1. Bean WJ,
    2. Schell M,
    3. Katz J,
    4. Kawaoka Y,
    5. Naeve C,
    6. Gorman O,
    7. Webster RG
    . 1992. Evolution of the H3 influenza virus hemagglutinin from human and nonhuman hosts. J Virol66:1129–1138.
    OpenUrlAbstract/FREE Full Text
  59. 59.↵
    1. Both GW,
    2. Sleigh MJ,
    3. Cox NJ,
    4. Kendal AP
    . 1983. Antigenic drift in influenza virus H3 hemagglutinin from 1968 to 1980: multiple evolutionary pathways and sequential amino acid changes at key antigenic sites. J Virol48:52–60.
    OpenUrlAbstract/FREE Full Text
  60. 60.↵
    1. Bush RM,
    2. Fitch WM,
    3. Bender CA,
    4. Cox NJ
    . 1999. Positive selection on the H3 hemagglutinin gene of human influenza virus A. Mol Biol Evol16:1457–1465. doi:10.1093/oxfordjournals.molbev.a026057.
    OpenUrlCrossRefPubMedWeb of Science
  61. 61.↵
    1. Epperson S,
    2. Jhung M,
    3. Richards S,
    4. Quinlisk P,
    5. Ball L,
    6. Moll M,
    7. Boulton R,
    8. Haddy L,
    9. Biggerstaff M,
    10. Brammer L,
    11. Trock S,
    12. Burns E,
    13. Gomez T,
    14. Wong KK,
    15. Katz J,
    16. Lindstrom S,
    17. Klimov A,
    18. Bresee JS,
    19. Jernigan DB,
    20. Cox N,
    21. Finelli L
    . 2013. Human infections with influenza A(H3N2) variant virus in the United States, 2011-2012. Clin Infect Dis57(Suppl 1):S4–S11. doi:10.1093/cid/cit272.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. Larson LJ,
    2. Henningson J,
    3. Sharp P,
    4. Thiel B,
    5. Deshpande MS,
    6. Davis T,
    7. Jayappa H,
    8. Wasmoen T,
    9. Lakshmanan N,
    10. Schultz RD
    . 2011. Efficacy of the canine influenza virus H3N8 vaccine to decrease severity of clinical disease after cochallenge with canine influenza virus and Streptococcus equi subsp. zooepidemicus. Clin Vaccine Immunol18:559–564. doi:10.1128/CVI.00500-10.
    OpenUrlAbstract/FREE Full Text
  63. 63.↵
    1. Hale BG,
    2. Randall RE,
    3. Ortin J,
    4. Jackson D
    . 2008. The multifunctional NS1 protein of influenza A viruses. J Gen Virol89:2359–2376. doi:10.1099/vir.0.2008/004606-0.
    OpenUrlCrossRefPubMedWeb of Science
  64. 64.↵
    1. Baskin CR,
    2. Bielefeldt-Ohmann H,
    3. Garcia-Sastre A,
    4. Tumpey TM,
    5. Van Hoeven N,
    6. Carter VS,
    7. Thomas MJ,
    8. Proll S,
    9. Solorzano A,
    10. Billharz R,
    11. Fornek JL,
    12. Thomas S,
    13. Chen CH,
    14. Clark EA,
    15. Murali-Krishna K,
    16. Katze MG
    . 2007. Functional genomic and serological analysis of the protective immune response resulting from vaccination of macaques with an NS1-truncated influenza virus. J Virol81:11817–11827. doi:10.1128/JVI.00590-07.
    OpenUrlAbstract/FREE Full Text
  65. 65.↵
    1. Choi EH,
    2. Song MS,
    3. Park SJ,
    4. Pascua PN,
    5. Baek YH,
    6. Kwon HI,
    7. Kim EH,
    8. Kim S,
    9. Jang HK,
    10. Poo H,
    11. Kim CJ,
    12. Choi YK
    . 2015. Development of a dual-protective live attenuated vaccine against H5N1 and H9N2 avian influenza viruses by modifying the NS1 gene. Arch Virol160:1729–1740. doi:10.1007/s00705-015-2442-y.
    OpenUrlCrossRef
  66. 66.↵
    1. Pica N,
    2. Langlois RA,
    3. Krammer F,
    4. Margine I,
    5. Palese P
    . 2012. NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol86:10293–10301. doi:10.1128/JVI.01131-12.
    OpenUrlAbstract/FREE Full Text
  67. 67.↵
    1. Quinlivan M,
    2. Zamarin D,
    3. Garcia-Sastre A,
    4. Cullinane A,
    5. Chambers T,
    6. Palese P
    . 2005. Attenuation of equine influenza viruses through truncations of the NS1 protein. J Virol79:8431–8439. doi:10.1128/JVI.79.13.8431-8439.2005.
    OpenUrlAbstract/FREE Full Text
  68. 68.↵
    1. Richt JA,
    2. Lekcharoensuk P,
    3. Lager KM,
    4. Vincent AL,
    5. Loiacono CM,
    6. Janke BH,
    7. Wu WH,
    8. Yoon KJ,
    9. Webby RJ,
    10. Solorzano A,
    11. Garcia-Sastre A
    . 2006. Vaccination of pigs against swine influenza viruses by using an NS1-truncated modified live-virus vaccine. J Virol80:11009–11018. doi:10.1128/JVI.00787-06.
    OpenUrlAbstract/FREE Full Text
  69. 69.↵
    1. Solorzano A,
    2. Webby RJ,
    3. Lager KM,
    4. Janke BH,
    5. Garcia-Sastre A,
    6. Richt JA
    . 2005. Mutations in the NS1 protein of swine influenza virus impair anti-interferon activity and confer attenuation in pigs. J Virol79:7535–7543. doi:10.1128/JVI.79.12.7535-7543.2005.
    OpenUrlAbstract/FREE Full Text
  70. 70.↵
    1. Steel J,
    2. Lowen AC,
    3. Pena L,
    4. Angel M,
    5. Solorzano A,
    6. Albrecht R,
    7. Perez DR,
    8. Garcia-Sastre A,
    9. Palese P
    . 2009. Live attenuated influenza viruses containing NS1 truncations as vaccine candidates against H5N1 highly pathogenic avian influenza. J Virol83:1742–1753. doi:10.1128/JVI.01920-08.
    OpenUrlAbstract/FREE Full Text
  71. 71.↵
    1. Vincent AL,
    2. Ma W,
    3. Lager KM,
    4. Janke BH,
    5. Webby RJ,
    6. Garcia-Sastre A,
    7. Richt JA
    . 2007. Efficacy of intranasal administration of a truncated NS1 modified live influenza virus vaccine in swine. Vaccine25:7999–8009. doi:10.1016/j.vaccine.2007.09.019.
    OpenUrlCrossRefPubMedWeb of Science
  72. 72.↵
    1. Feng KH,
    2. Gonzalez G,
    3. Deng L,
    4. Yu H,
    5. Tse VL,
    6. Huang L,
    7. Huang K,
    8. Wasik BR,
    9. Zhou B,
    10. Wentworth DE,
    11. Holmes EC,
    12. Chen X,
    13. Varki A,
    14. Murcia PR,
    15. Parrish CR
    . 2015. Equine and canine influenza H3N8 viruses show minimal biological differences despite phylogenetic divergence. J Virol89:6860–6873. doi:10.1128/JVI.00521-15.
    OpenUrlAbstract/FREE Full Text
  73. 73.↵
    1. Murakami S,
    2. Horimoto T,
    3. Yamada S,
    4. Kakugawa S,
    5. Goto H,
    6. Kawaoka Y
    . 2008. Establishment of canine RNA polymerase I-driven reverse genetics for influenza A virus: its application for H5N1 vaccine production. J Virol82:1605–1609. doi:10.1128/JVI.01876-07.
    OpenUrlAbstract/FREE Full Text
  74. 74.↵
    1. Cheng BY,
    2. Ortiz-Riano E,
    3. Nogales A,
    4. de la Torre JC,
    5. Martinez-Sobrido L
    . 2015. Development of live-attenuated arenavirus vaccines based on codon deoptimization. J Virol89:3523–3533. doi:10.1128/JVI.03401-14.
    OpenUrlAbstract/FREE Full Text
  75. 75.↵
    1. Nogales A,
    2. Baker SF,
    3. Martinez-Sobrido L
    . 2014. Replication-competent influenza A viruses expressing a red fluorescent protein. Virology476C:206–216.
    OpenUrlCrossRef
  76. 76.↵
    National Research Council, Committee for the Update of the Guide for the Care and Use of Laboratory Animals, Institute for Laboratory Animal Research, and National Academies Press. 2011. Guide for the care and use of laboratory animals, 8th ed. National Academies Press, Washington, DC.
PreviousNext
Back to top
Download PDF
Citation Tools
Temperature-Sensitive Live-Attenuated Canine Influenza Virus H3N8 Vaccine
Aitor Nogales, Laura Rodriguez, Caroline Chauché, Kai Huang, Emma C. Reilly, David J. Topham, Pablo R. Murcia, Colin R. Parrish, Luis Martínez-Sobrido
Journal of Virology Jan 2017, 91 (4) e02211-16; DOI: 10.1128/JVI.02211-16

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Journal of Virology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Temperature-Sensitive Live-Attenuated Canine Influenza Virus H3N8 Vaccine
(Your Name) has forwarded a page to you from Journal of Virology
(Your Name) thought you would be interested in this article in Journal of Virology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Temperature-Sensitive Live-Attenuated Canine Influenza Virus H3N8 Vaccine
Aitor Nogales, Laura Rodriguez, Caroline Chauché, Kai Huang, Emma C. Reilly, David J. Topham, Pablo R. Murcia, Colin R. Parrish, Luis Martínez-Sobrido
Journal of Virology Jan 2017, 91 (4) e02211-16; DOI: 10.1128/JVI.02211-16
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • INTRODUCTION
    • RESULTS
    • DISCUSSION
    • MATERIALS AND METHODS
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

Dog Diseases
Influenza A Virus, H3N8 Subtype
influenza vaccines
Orthomyxoviridae Infections
Vaccines, Attenuated
influenza
influenza vaccines

Related Articles

Cited By...

About

  • About JVI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #Jvirology

@ASMicrobiology

       

 

JVI in collaboration with

American Society for Virology

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0022-538X; Online ISSN: 1098-5514