Research Article
1 July 2013

Genetic Requirement for Hemagglutinin Glycosylation and Its Implications for Influenza A H1N1 Virus Evolution

ABSTRACT

Influenza A virus has evolved and thrived in human populations. Since the 1918 influenza A pandemic, human H1N1 viruses had acquired additional N-linked glycosylation (NLG) sites within the globular head region of hemagglutinin (HA) until the NLG-free HA head pattern of the 1918 H1N1 virus was renewed with the swine-derived 2009 pandemic H1N1 virus. Moreover, the HA of the 2009 H1N1 virus appeared to be antigenically related to that of the 1918 H1N1 virus. Hence, it is possible that descendants of the 2009 H1N1 virus might recapitulate the acquisition of HA head glycosylation sites through their evolutionary drift as a means to evade preexisting immunity. We evaluate here the evolution signature of glycosylations found in the globular head region of H1 HA in order to determine their impact in the virulence and transmission of H1N1 viruses. We identified a polymorphism at HA residue 147 associated with the acquisition of glycosylation at residues 144 and 172. By in vitro and in vivo analyses using mutant viruses, we also found that the polymorphism at HA residue 147 compensated for the loss of replication, virulence, and transmissibility associated with the presence of the N-linked glycans. Our findings suggest that the polymorphism in H1 HA at position 147 modulates viral fitness by buffering the constraints caused by N-linked glycans and provide insights into the evolution dynamics of influenza viruses with implications in vaccine immunogenicity.

INTRODUCTION

Influenza A virus (IAV) causes a serious public health problem. The virus is endemic in the human population and circulates globally, causing seasonal epidemics that result in the deaths of up to half a million people each year (1). Occasionally, a large antigenic shift occurs in IAV, resulting in an influenza pandemic, which can threaten the lives of millions (2). The first pandemic of the 21st century was caused by an H1N1 subtype of IAV in 2009 (2009 H1N1) (3, 4). The hemagglutinin (HA) of this virus was antigenically distinct from the previously circulating seasonal human H1N1 (hH1N1) viruses (57) and resembled that of swine-lineage H1N1 viruses (8). Based on a structural analysis, the HA epitopes of the 2009 H1N1 virus appeared to be similar to those of the 1918 H1N1 “Spanish Flu” virus, which caused one of the largest known pandemics and is the evolutionary ancestor of human- and swine-lineage H1N1 viruses (911). Another similarity between the two pandemic viruses is the absence of N-linked glycosylation (NLG) at the globular head of HA (12, 13). These features of the 2009 H1N1 virus explained why antibodies that protected against infection with the 2009 H1N1 virus were detected in the elderly who had experienced the 1918 pandemic and in people who had been vaccinated against the “swine flu” in 1976 (5, 6, 14).
During evolution in humans, IAV accumulates genetic mutations at the antigenic sites of HA that circumvent preexisting immunity, a process known as antigenic drift (15). Some of these mutations result in variations in the glycosylation state of the HA associated with changes in viral antigenicity (16) and with immune evasion (17). Since 1918, hH1N1 viruses have acquired multiple NLGs, which occur at asparagine (N) residues in accessible N-Xaa-S/T (Xaa, any amino acid except proline) sequons, in the top, side, and stem regions of HA (12, 18). However, HA is neither hyperglycosylated (19) nor glycosylated at random sites (20). Using the HA amino acid sequences of hH1N1 viruses that were available from influenza virus database of the National Centers for Biotechnology Information (NCBI), we compared the NLG patterns in the head region of these HAs. As noted previously (12), we found that various combinations of NLG using only five glycosites (asparagines at 142, 144, 172, 177, and 179 of HA; H1 numbering was applied in the present study unless otherwise specified) have been utilized by hH1N1 viruses (Table 1). In the 1930s, a single NLG at residues 142, 144, or 179 was placed in the HA of hH1N1 viruses. From 1942 to 1985, hH1N1 viruses maintained several combinations of NLGs at residues 144, 172, 177, and/or 179; residue 179 was utilized mostly until 1948. In 1986, hH1N1 viruses exhibited a double mutation, K142N and N144S/T, which allowed a glycan shift from residue 144 to residue 142. The NLGs at residues 142 and 177 then became the NLG signature of hH1N1 viruses until the sugar-free HA head was observed in the 2009 H1N1 viruses (Table 1) (21).
Table 1
Table 1 Combinations of NLG sites and the genetic signature at residue 147 of representative HA sequences
Virus GenBank no. Residues harboring N-linked glycans and amino acids from residue 147 at the globular head of HAa
142 144 147 172 177 179
A/South Carolina/1/1918 AAD17229     K      
A/Wilson-Smith/33 ABD77796     K     o
A/WSN/33 AAA43209 o   Absent      
A/Puerto Rico/8/34 ACF41834     Absent      
A/Melbourne/35 ABD62781   o K      
A/Henry/36 ABO38351     I      
A/Hickox/40 ABI20826     I     o
A/Bellamy/42 ABD62843   o K     o
A/Weiss/43 ABD79101     R      
A/AA/Marton/43 ABO38054   o R     o
A/Iowa/43 ABO38373     Absent     o
A/USA/L3/47 AEM23889     R o o  
A/Albany/4835/48 ABN59401   o R o   o
A/Hemsbury/48 ADT79097   o R      
A/Netherlands/002K1/49 ADT78929   o R   o  
A/Albany/4836/50 ABP49316   o R o    
A/Fort Warren/1/50 ABD61735     R o    
A/Albany/12/51 ABP49481   o R o o  
A/Netherlands/001B1/56 ADT78859 o   R o o  
A/Christ's Hospital/157/82 ABO52797   o K o    
A/Chile/1/83 ABO38340   o K o o  
A/Tonga/14/84 ABO38406   o K   o  
A/Singapore/6/86 ABO38395 o   K o o  
A/Taiwan/1/86 ABF21274 o   K o    
A/Siena/4/87 ACV49666     K o o  
A/Siena/9/89 ACL12261 o   K   o  
A/Beijing/262/95 ACF41867 o   Absent   o  
A/Ohio/01/2007 ACQ73203     R     o
A/California/04/2009 ACP41105     K      
a
K, lysine; I, isoleucine; R, arginine; o, NLG attachment at defined residues; Absent, residue 147 was absent in the HA of virus.
A recent report showed that the presence of lysine (K) at HA residue 147 (K147; referred to as 133a in H3 numbering) of the A/California/04/2009 (Ca/04; a 2009 H1N1 virus) is associated with the stabilization of the interaction between HA and sialic acid (SA) (22). K147 is also present in the HA of the 1918 H1N1 virus; however, this signature was not present in all of the subsequent hH1N1 descendants. Some of the 1918 H1N1 descendants had an arginine (R) or isoleucine (I) at HA residue 147 instead of a K (referred to as HA-147-bearing hH1N1 viruses), whereas in others this amino acid was even absent (referred to as HA-Δ147 hH1N1 viruses) (Table 1).
Based on these phenomena, we hypothesized that the NLG-free headed and K147-bearing HA of 2009 H1N1 virus might evolve into a similar genetic flow as previously seen in 1918 H1N1 descendant viruses. We thus investigated how the attached N-linked glycans and the polymorphism at HA residue 147 affected viral characteristics. Using mutant viruses generated by reverse genetics, we show here the importance of the polymorphism at HA residue 147. When the globular head region of HA acquired specific N-linked glycans, HA residue 147 was indispensable for viral fitness, and it allowed NLG-containing viruses to be transmissible in an animal model. In addition, the HA residue 147 affected the immunogenicity of the H1N1 viruses. Considered together, we propose that the polymorphism at HA residue 147 cooperates with NLG acquisition at the HA head of hH1N1 viruses to evade preexisting immunity without compromising viral fitness to the point of losing their transmissibility.

MATERIALS AND METHODS

Viruses and cells.

Viruses were propagated in 10-day-old embryonated chicken eggs. To determine the virus titers, the viruses were hemagglutinated with turkey red blood cells (tRBCs), or plaque assays were carried out in Madin-Darby canine kidney (MDCK) cells. All of the viruses were plaque purified and sequenced before use in the experiments. MDCK cells were obtained from the American Type Culture Collection (ATCC), maintained in Eagle minimal essential medium, and used for the cell-based assay. Human embryonic kidney (293T) cells and human lung epithelial (A549) cells were obtained from the ATCC and maintained in Dulbecco modified Eagle medium. All of the media were supplemented with 10% fetal bovine serum and antibiotics.

Animal experiments.

To minimize animal suffering, all animal procedures performed in the present study were conducted in accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the Animal, Plant, and Fisheries Quarantine and Inspection Agency of Korea. The protocol was approved by the Institutional Animal Care and Use Committee of Hallym University (permit number Hallym 2012-24).
To determine the 50% lethal dose (LD50) for mice and their body weight changes, five C57BL/6 mice (female, 5 weeks old; NaraBiotech, Korea) per group were anesthetized and intranasally infected with each virus. Body weight changes and survival rates were recorded until 14 days postinfection (dpi). Mice that experienced more than a 25% loss in body weight were considered experimentally dead and were humanely euthanized. The LD50 was determined using the Reed and Muench method (23). To determine virus titers in the lungs, six C57BL/6 mice per group were anesthetized and intranasally infected with 105 PFU of each virus. At 3 and 6 dpi, three mice per group were sacrificed for lung collection. The collected lung samples were homogenized with TissueLyzer II (Qiagen, Germany) and were titrated to assess the presence of virus by plaque assay in MDCK cells.
For a direct contact transmission model, guinea pigs (Hartley strain, female, 5 to 6 weeks old; Charles River Laboratories, Wilmington, MA) were anesthetized and intranasally infected with 105 PFU of each virus. The following day, a naive guinea pig was co-caged with each infected guinea pig. To collect nasal wash samples, the anesthetized guinea pigs were intranasally lavaged with 1 ml of phosphate-buffered saline supplemented with penicillin-streptomycin and 0.3% bovine serum albumin (PBS-PS-BSA), and the lavage fluid was allowed to drain onto a sterilized petri dish. The virus that was present in the nasal wash samples was titrated by plaque assay in MDCK cells.

Antibodies.

To obtain guinea pig polyclonal antibodies specific for each virus, naive (influenza virus-negative) guinea pigs were intranasally primed and boosted with 105 PFU of virus at 2-week intervals. Antisera were then prepared from whole blood and treated with RDE (neuraminidase from Vibrio cholerae; Denka Seiken, Japan) before use. Sheep polyclonal antibodies specific for 2009 H1N1 HA (influenza anti-A/California/7/2009 H1N1 HA serum, NIBSC code 11/110) were obtained from the NIBSC (Hertfordshire, United Kingdom) and were used in Western blots to detect HA proteins.

Rescue of rK/09 and HA mutant viruses.

The A/Korea/01/2009 (K/09; NCBI taxonomy ID 644289) virus is a 2009 H1N1 strain isolated in Korea. This virus was used as the backbone virus for reverse genetics in the present study. All eight gene segments of the K/09 virus were cloned into ambisense pDZ plasmids (kindly provided by Peter Palese, Mount Sinai School of Medicine, New York, NY). After sequence analysis of these plasmids, HA mutant plasmids were constructed to reflect various NLG statuses and the genetic polymorphism at residue 147. The rK/09 and HA mutant viruses were then rescued by reverse genetics as previously described (24). Briefly, cocultured 293T/MDCK cells were transfected with eight pDZ plasmids encoding the genes and proteins of wild-type K/09 or HA mutant viruses. At 48 to 72 h after transfection, supernatants were inoculated into 10-day-old embryonated chicken eggs and maintained at 37°C. The allantoic fluids were harvested 48 to 72 h later and titrated in HA and plaque assays to determine the viral presence. The rescued viruses were purified with a plaque assay and then repropagated in 10-day-old embryonated chicken eggs. All of the rescued viruses were sequence confirmed before being used in the experiments.

RBC binding assay.

To evaluate the RBC binding avidities of the HAs in each virus, a protocol modified from the work of Hensley et al. (25) was used. Briefly, turkey RBCs (tRBCs) or human RBCs (hRBCs) (turkey or human blood in Alsever's solution, Rockland, PA) were treated with 0.025 to 3.2 μg of sialidase (α2-3,6,8 neuraminidase from Vibrio cholerae; Roche, Germany)/ml at 37°C. After 1 h, the sialidase-treated RBCs were washed and diluted to 0.5% (vol/vol) in the RBC solution with PBS. A total of four HA units of virus in 50 μl were then allowed to agglutinate with RBCs for another 30 min. The HA-RBC agglutination was read, and the RBC binding avidities of viruses were determined in three independent experiments.

Growth kinetics in cultured cells.

For the growth property analysis, the MDCK and A549 cell monolayers were inoculated with each virus at a multiplicity of infection (MOI) of 0.001. After 1 h, the cells were washed five times and maintained in the appropriate medium supplemented with 0.3% BSA in the presence of TPCK (tolylsulfonyl phenylalanyl chloromethyl ketone)-treated trypsin (1 μg/ml). At the indicated time points, the supernatants were collected and titrated for viral presence by plaque assay in the MDCK cells. The results are presented as the mean titers at each time point of more than three independent experiments.

HI assay.

In 96-well plates, the hemagglutination inhibition (HI) titers of guinea pig antisera were determined against homologous and heterologous viruses using tRBCs. Briefly, 8 HA units of viruses in 25 μl were incubated with the same amount of guinea pig antisera at 37°C. After 1 h of incubation, the virus and each antiserum mixture were agglutinated with 50 μl of 0.5% tRBC for 30 min. The HI titers of antisera against homologous and heterologous viruses were determined in three independent experiments.

Plaque reduction neutralization test (PRNT).

A total of 100 PFU of each virus was incubated with the same amount of guinea pig antisera at 37°C. After 1 h of incubation, the virus and each antiserum mixture were plaque assayed in the MDCK cells. The control wells of MDCK cells were infected only with virus. The neutralization titers were determined by the dilution of each antiserum that resulted in a 50% reduction in the plaque titers compared to the controls. The PRNT titers of each antiserum were determined in three independent experiments.

Western blotting and PNGase F treatment.

To confirm the biochemical utilization of NLG in the HA of the rK/09 and HA mutant viruses, the monolayers of MDCK cells were infected with an MOI of 3. After 16 h of infection, the cell lysates were prepared for SDS-PAGE. The HA in the cell lysates was primarily detected by sheep anti-2009 pH1N1 HA-specific polyclonal antibodies and then secondarily detected by anti-sheep IgG-HRP antibodies. To confirm the mobility identities in the HAs of the rK/09 and HA mutant viruses (after introduced N-linked glycans were removed), the prepared cell lysates were incubated with PNGase F (from Flavobacterium meningosepticum, recombinant from E. coli; Roche, Germany) at 37°C for 1 h, and the HAs were then detected using the procedures described above.

RESULTS

Sequence and structural analysis.

We first analyzed amino acid changes at HA residue 147 during the evolution of human H1 HAs and its relationship to NLG patterns in the globular head region of HA. In the HAs of early 1918 H1N1 descendants until the late 1940s, residue 147 was highly polymorphic and was either absent (Δ147) or occupied by an amino acid K, R, or I (Fig. 1 and see Table S1 in the supplemental material). However, R147 became the prevalent polymorphism between 1948 and 1956 and had been a dominant signature until 1981 since the reintroduction of hH1N1 viruses in 1976. Then, R147 was succeeded by another polymorphic signature, K147, throughout 1980s and early 1990s until the sudden reappearance of HA-Δ147 variant was reported with the A/Beijing/262/95 (Bj/95) virus in 1995. Soon, this variant claimed dominance over HA-147-bearing viruses, and residue 147 became no longer obligatory in the HA of seasonal H1N1 viruses (Fig. 1 and see Table S1 in the supplemental material). In NLG pattern analysis, it was revealed that the HA-Δ147 hH1N1 viruses were never glycosylated at residues 144 and 172 (Table 2), whereas these glycosylation sites were found in HA-147-bearing hH1N1 viruses (Table 1). These observations suggest that the presence of HA residue 147 is required to accommodate 144 and 172 NLG sites.
Fig 1
Fig 1 Polymorphic signatures at HA residue 147 of hH1N1 viruses. Since 1918, the HA residue 147 was alternatively occupied by two amino acids, lysine (K) or arginine (R) in most of hH1N1 viruses. However, this residue was absent (△) in some other hH1N1 viruses. Isoleucine (I) at HA 147 only identified until 1940 was excluded in this figure.
Table 2
Table 2 NLG patterns at the HA globular head region of Δ147 hH1N1 representative viruses
Virus representative GenBank no. Residues harboring N-linked glycans at the globular head of HA in the HA-Δ147 hH1N1 virusesa
142 144 147 172 177 179
A/WSN/33 AAA43209 o   Absent      
A/Puerto Rico/8/34 ACF41834     Absent      
A/Iowa/43 ABO38373     Absent     o
A/Mongolia/153/88 CAA91081     Absent      
A/Mongolia/111/91 CAA91082     Absent      
A/Beijing/262/95 ACF41867 o   Absent   o  
A/New Caledonia/20/99 ACF41878 o   Absent   o  
A/Thailand/Siriraj-07/2000 ABS76427 o   Absent      
A/New York/205/2001 AAZ38627 o   Absent   o  
A/Solomon Islands/3/2006 ABU50586 o   Absent   o  
A/Brisbane/59/2007 ADE28750 o   Absent   o  
a
See Table 1, footnote a.
We next analyzed the location of HA residue 147 and of NLG sites in the three-dimensional structure of the HA head. We used the already known X-ray structures of the HA of A/Puerto Rico/8/34 (PR8) (26), in which residue 147 is absent, and of the HA of Ca/04 (9), which has K147. We found that residues 144 and 172, which correspond to the NGL sites of some of the H1N1 strains but are not glycosylated in PR8 and Ca/04, are located adjacent to the HA receptor binding site (RBS) in the PR8 HA (Fig. 2A and B). In contrast, residue 147 in the HA of the Ca/04 virus pushes away residues 144 and 172 from the RBS, as the lysine side chain of residue 147 forms a ridge at the edge of the RBS (Fig. 2C and D). K147, together with two other adjacent lysine residues K159 and K236, may generate a lysine “fence” (27) that protects RBS-SA binding from steric hindrance in the case of glycosylation at residues 144 and/or 172.
Fig 2
Fig 2 Potential NLG sites at the globular head region of K147-bearing and Δ147 H1 HA crystal structures. Using PyMOL software, the potential NLG sites of hH1N1 viruses are indicated by color and residue number (H1 numbering) in the vicinity of HA RBS. (A and B) Using the Δ147 HA structure of the PR8 virus (PDB ID 1RU7), the close proximity of RBS (yellow) to NLG sites 144 and 172 (red) was highlighted by magnified three-dimensional representations. (C and D) In contrast, the side chain of K147 (blue) constituted a “fence” between the receptor-binding site and 144 and 172 in the HA of the 2009 pH1N1 virus (structure of A/California/04/09; PDB ID 3LZG).

Generation and characterization of mutant viruses.

On the basis of the above sequence analyses and structural predictions, we constructed seven plasmids, each carrying a mutant HA from a prototypic 2009 H1N1 virus, A/Korea/01/09 (K/09) (28) containing different combinations of NLG (142, 144, 172, and/or 177; NLG 179 was excluded because it was less frequently utilized) and residue 147 (K, R, or ΔK147; I was excluded because it was less frequently adopted) (Table 3). This allowed us to examine whether the NLG patterns and the genetic polymorphism of HA residue 147 found in previous seasonal H1N1 influenza viruses affect the virulence, transmissibility, and immunogenicity of hH1N1 viruses. Mutant viruses were then generated by reverse genetics on the backbone of K/09. Mutations in each HA were confirmed by sequence analysis, and no other variations were found except for the intended mutations within the HA.
Table 3
Table 3 Construction of rK/09 HA mutant plasmids reflecting the NLG status and amino acid at residue 147 of the globular head of HA in hH1N1 viral representatives
Plasmid Rescued virus Virus representative Residues harboring N-linked glycans and amino acids from residue 147 at the globular head of HAc
142 144 147 172 177
pK/09:HA rK/09 A/Korea/01/2009     K    
pΔK147 rΔK147 A/Puerto Rico/8/1934     Deleted    
p142 r142 a o   K    
p142-ΔK147 r142-ΔK147 A/WSN/1933 o   Deleted    
p144-R147-177 r144-R147-177 A/Netherlands/002K1/1949   o R   o
p144-R147-172-177 r144-R147-172-177b A/Brazil/11/1978   o R o o
p142-177 r142-177 A/Texas/36/1991 o   K   o
p142-ΔK147-177 r142-ΔK147-177 A/Beijing/262/1995 o   Deleted   o
a
–, There was no virus representative identified for the r142 virus, and it was used as a control.
b
The r144-R147-172-177 virus was not rescued from repeated trials.
c
See Table 1, footnote a.
We checked that the introduced NLGs in mutant viruses were utilized in MDCK cells. A shift in molecular weights was observed after treatment with PNGase F, to remove N-linked glycans, indicating that the HA of mutant viruses was glycosylated (Fig. 3A). We then evaluated the phenotypes of the viruses by plaque assay in MDCK cells. The r142 and r142-177 viruses produced large plaques that were almost identical in size to those of the parental rK/09 virus (Fig. 3B). For the r142-177 virus, there was heterogeneity in the plaque morphology, which may have been caused by the additional NLG at residue 177; however, most plaques exhibited similar phenotypes. A drastic transformation of the viral phenotype in plaques was observed for HA-ΔK147 viruses. Removal of K147 (rΔK147 virus) caused no changes in the plaque morphology compared to the rK/09 virus. However, the plaques decreased in size when a new NLG was introduced at residue 142 (r142-ΔK147 virus), and this size decrease continued further when a second NLG was added at residue 177 (r142-ΔK147-177 virus) (Fig. 3B). These findings indicate that the plaque phenotype of HA-ΔK147 viruses, but not HA-K147-bearing viruses, is markedly influenced by the introduction of N-linked glycans on the globular head of HA.
Fig 3
Fig 3 In vitro characterization of rK/09 and HA mutant viruses. (A) Confirmation of the utilization of NLG in the HA of rK/09 and HA mutant viruses by the Western blotting and PNGase F treatments. (B) Comparison of viral phenotypes by a plaque assay in MDCK cells. (C) Evaluation of cell binding avidity of viruses using sialidase-treated turkey and human RBCs in triplicate. The results are presented as mean values and standard deviations (SD). (D and E) Evaluation of viral growth properties in MDCK (D) and A549 (E) cells at an MOI of 0.001. The results are means and SD obtained from more than three independent titers at each time point. The detection limit of viral presence was 10 PFU/ml. The statistical significance of the differences in growth properties between rK/09 and HA mutant viruses was assessed by using a Student t test (*, P < 0.05; **, P < 0.01). Error bars denote the SD.
To address whether the attached NLG sites support or impede efficient HA-SA binding, we analyzed the avidity of binding of mutant viruses to tRBCs and hRBCs. In the HA-RBC binding assay, RBCs were treated with various concentrations of sialidase, which hydrolyzed the terminal SA linkage presented on the surfaces of RBCs. When agglutinated, the HA of the parental rK/09 virus could bind to tRBCs treated with up to 0.4 μg of sialidase/ml, and the HA of r142 and r142-177 viruses had a similar binding avidity (Fig. 3C). NLG at residue 142 alone or at residues 142 and177 did not affect the receptor binding avidity in the presence of HA-K147. In HA-ΔK147 viruses, however, the HA receptor binding ability was markedly reduced, indicating that the attached N-linked glycans affect the avidity of this interaction. A 0.2-μg/ml sialidase treatment was sufficient to prevent the HA-tRBC bridge formation by the r142-ΔK147 virus, and even less sialidase was needed to break the r142-ΔK147-177 virus-tRBC bridge (Fig. 3C). The same patterns of HA-RBC binding events were reproduced with hRBCs (Fig. 3C). These results suggest that HA residue 147 may have a critical role in the HA binding avidity of hH1N1 viruses. Only in the presence of the K147 residue could viruses maintain viral spreading in plaque assay and HA receptor binding avidity even under the possible constraints of additional NLG, whereas the HA-ΔK147 viruses were less functional in the presence of new NLG.
We then assessed the effects of NLG on the growth properties of viruses in two cell lines: MDCK and human lung epithelial (A549) cells. In MDCK cells, the introduction of NLG at the globular head of HA had marginal effects on the virus yields compared to the parental virus except for the r142-ΔK147-177 virus, which showed retarded growth until 48 h postinfection (Fig. 3D). In addition, the r142-ΔK147-177 virus, as well as the r142-ΔK147 virus, exhibited a greatly diminished growth in A549 cells, whereas the other viruses (except for r144-R147-177) replicated similarly to the parental virus (Fig. 3E). As seen in the plaque morphology and RBC binding assays, the growth properties of the HA-ΔK147 viruses, not those of the HA-K147-bearing viruses, were attenuated in A549 cells when NLG was attached in the head of HA.
Reduced RBC binding and growth properties were also observed for the r144-R147-177 virus (Fig. 3C and E). This virus exhibits one of the NLG patterns seen at the HA globular head of the hH1N1 viruses that circulated from the 1940s to the 1980s and thus contains arginine instead of lysine at HA residue 147. Although there is no direct evidence of the reduced fitness for the R147-bearing viruses, arginine at other positions has been known to modulate the HA-SA interaction (29). Steric hindrance and restructured hydrogen bonds have also been identified in relation to the guanidinium cation, a lengthy arginine side chain (30, 31), which might be attributable to the attenuated growth and RBC binding avidity of the r144-R147-177 virus. In addition, the more viral fitness should be considered the more NLG pressure is present around the RBS, which might be the reason why the r144-R147-172–177 virus could not be rescued in repeated trials (data not shown). Taken together, these findings suggest that the presence of residue 147 increases the viral capacity to incorporate additional NLG at the globular head of HA and that lysine (instead of arginine) may be a better signature to compensate for NLG-induced interference.

Pathogenesis of mutant viruses in mice.

Next, we assessed the effects of NLG and residue 147 of HA on viral pathogenesis in C57BL/6 mice (Table 4). As previously reported using other 2009 H1N1 pandemic viruses (14), the parental rK/09 virus replicated well in the lungs of mice (107.18 ± 0.39 PFU/ml and 106.59 ± 0.35 PFU/ml at 3 and 6 days postinfection, respectively) and resulted in a body weight loss of more than 20% in infected mice. A 50% mouse lethal dose (MLD50) of the rK/09 virus was determined to 105.67 PFU. However, none of the mutant viruses was as lethal (MLD50 > 106 PFU), even though comparable levels of viral replication were observed in the lungs of the infected mice for the r142, r142-177, and rΔK147 viruses. The r142-ΔK147, r142-ΔK147-177, and r144-R147-177 viruses replicated at lower levels in the lungs than did the parent virus, and mice infected with these mutant viruses did not experience weight loss (Table 4). Considering these findings together, the introduction of NLG at the globular head of HA results in different viral pathogenicities in mice according to the genetic signature of HA residue 147.
Table 4
Table 4 Pathological findings in C57BL/6 mice infected with rK/09 and HA mutant virusesa
Virus MLD50 % wt change ± SD (dpi)b Virus titer (log10[(PFU/ml/g) ± SD]) in lungs
3 dpi 6 dpi
Mock   ↑13.87 ± 2.60 (13) NDc ND
rK/09 105.67 ↓20.33 ± 1.69 (8) 7.18 ± 0.39 6.59 ± 0.35
rΔK147 >106 ↓2.23 ± 4.41 (6)** 7.33 ± 0.17 6.59 ± 0.30
r142 >106 ↓5.69 ± 3.56 (7)** 7.44 ± 0.26 7.27 ± 0.12
r142-ΔK147 >106 ↑17.46 ± 3.41 (14)** 6.48 ± 0.16* 6.01 ± 0.14
r142-177 >106 ↓7.01 ± 2.49 (7)** 6.97 ± 0.23 6.88 ± 0.21
r142-ΔK147-177 >106 ↑20.88 ± 5.64 (13)** 6.87 ± 0.18** 6.71 ± 0.11
r144-R147-177 >106 ↑12.68 ± 3.28 (13)** 5.90 ± 0.21** 5.76 ± 0.24*
a
*, P < 0.05; **, P < 0.01.
b
The weight change of individual groups was determined as the maximum percent difference in the body weight of mice during the 14 days of observation after infection (up arrow [↑], increase after infection; down arrow [↓], decrease after infection). dpi, days postinfection.
c
ND, not detected.

Transmission of mutant viruses in guinea pigs.

Transmissibility is a prerequisite for IAV to spread between individuals and to perpetuate itself in hosts (32, 33). The receptor specificity of HA has been also considered one of the key genetic determinants for the inter- and intraspecies transmission of IAV (30, 3437). However, the mechanism by which N-linked glycans at the globular head of HA modulate the transmission of IAV remains to be defined. In addition, the genetic polymorphism of HA residue 147 seems to be an important determinant of virulence. Thus, we evaluated the transmission of HA mutant viruses from one individual to another, using a guinea pig model validated for influenza virus transmission studies (38) (Fig. 4). In four of six uninfected guinea pigs that were exposed to infected guinea pigs, the parental rK/09 virus was transmitted by direct contact, as shown by the shedding of virus into the nasal respiratory tracts (4/6) (Fig. 4B). For the r142 and r142-177 viruses, six and five of six initially uninfected, exposed guinea pigs became infected with the respective virus (6/6 and 5/6, respectively) (Fig. 4C and D). The HA-K147-bearing viruses were therefore able to infect guinea pigs and were successfully transmitted to exposed naive guinea pigs despite the introduction of NLG(s) at the HA head. In contrast, only the rΔK147 virus among the HA-ΔK147 viruses was transmissible to exposed guinea pigs (2/3) (Fig. 4F). The r142-ΔK147 virus was detected at low titers in the infected guinea pigs but was not detected in the nasal wash samples of the exposed guinea pigs (0/6) (Fig. 4G), and the r142-ΔK147-177 virus could not even infect guinea pigs and therefore was never transmissible to guinea pigs (0/6) (Fig. 4H). With regard to the r144-R147-177 virus, the infected guinea pigs exhibited relatively low titers of viral presence, and the virus was transmitted by direct contact in only one of three guinea pigs (1/3) (Fig. 4E). Based on these findings, the genetic signature of HA residue 147 is critical for the transmission of 2009 H1N1 virus in guinea pigs when NLG is introduced at the globular head of HA.
Fig 4
Fig 4 The transmission efficiency of rK/09 and HA mutant viruses in guinea pigs. (A to H) To evaluate the transmissibility of the rK/09 and HA mutant viruses, guinea pigs were infected intranasally with 105 PFU of each virus as indicated. The following day, a naive guinea pig (open symbols) was cocaged with the infected guinea pigs (solid symbols) to allow for direct contact. The viral infection and transmission were determined by plaque assay in MDCK cells using nasal wash samples collected from the guinea pigs every other day after the infection or exposure. The guinea pigs in the mock group were inoculated with PBS. The detection limit of the viral presence was 10 PFU/ml. The number of transmission/infection guinea pig sets for each virus is presented in parenthesis.

HI and neutralization reactivities of guinea pig sera.

To investigate whether the NLG and residue 147 status of HA affected the immunogenicity of mutant viruses, we then carried out a HI assay, which utilized guinea pig antisera raised by intranasal inoculation with each mutant virus. Each antiserum exhibited HI titers of 320 to 2,560 to the homologous virus (Table 5). In our results, the K147-bearing viruses induced higher levels of reactive HI antibodies than did their HA-ΔK147 NLG counterparts (rK/09 > rΔK147; r142 > r142-ΔK147; and r142-177 > r142-ΔK147-177 [see Table 5]). This better immunogenicity of the K147-bearing viruses was easily appreciated when HI titers were expressed in fold difference normalized to each homologous HI titer to remove the bias that could be caused by the absolute amounts of antibodies in each antiserum (Fig. 5A to G). In particular, the r142-177 virus induced antibodies that were the most cross-reactive in HI assays against all of the tested viruses. This cross-reactivity of the r142-177 antiserum was suggested using the 1918 HA (12) and was also confirmed in our study. In a plaque reduction neutralization test (PRNT), the r142-177 antiserum neutralized the plaques generated by other mutant viruses, at titers of 320 to 1,280 (Table 6), which indicated the same or a greater neutralization efficacy compared to the homologous virus (Fig. 5C). However, we found that the r142-ΔK147-177 virus, of which HA had the same NLG pattern with the 142-177 virus but K147 was deleted, was poorly immunogenic and produced only 10 and 20 titers of reactive HI antibodies against the r142 and r142-177 viruses, respectively (Fig. 5G and Table 5). The low neutralization efficacy of the r142-ΔK147-177 antiserum was also observed in PRNT assays to other viruses (Fig. 5G and Table 6). In addition to its role in the stabilization of the HA-SA interaction and the viral characteristics discussed above, the HA residue 147 may be an important factor for the immunogenic performance of the H1N1 viruses.
Table 5
Table 5 Hemagglutination inhibition titers of guinea pig antisera to homologous and heterologous viruses
Virus HI titers of guinea pig antiseraa
K/09 ΔK147 142-K147 142-ΔK147 142-K147-177 142-ΔK147-177 144-R147-177
rK/09 1,280 640 160–320 20 640 20 160
rΔK147 1,280–2,560 1,280 160–320 20 640 40 320
r142-K147 1,280–2,560 320 1,280–2,560 20 640 10 80
r142-ΔK147 2,560 2,560 1,280 320 1,280 2,560 640
r142-K147-177 640 160 320 10 640 20 40
r142-ΔK147-177 320 160 160 40–80 80 320 40
r144-R147-177 2,560 1,280–2,560 640–1,280 160–320 640–1,280 1,280–2,560 640
a
Single values indicate the same HI titers were obtained in three independent experiments. Ranged values indicate that different HI titers were obtained in three independent experiments.
Fig 5
Fig 5 Immunogenicity of the rK/09 and HA mutant viruses. (A to G) To investigate the serologic reactivities to homologous and heterologous viruses, guinea pig antisera that were raised against individual viruses were used for the HI assay (black bars) and PRNT (white bars) in MDCK cells. The HI and PRNT titers are mean values in triplicate. To remove the bias that resulted from the absolute quantity of polyclonal antibodies in the guinea pig antisera, the relative fold differences of HI and PRNT titers were calculated for the homologous titers: fold difference = log2[heterologous titer] – log2[homologous titer]. Error bars denote the SD.
Table 6
Table 6 Neutralization titers of guinea pig antisera to homologous and heterologous viruses
Virus Neutralization titers of guinea pig antiseraa
K/09 ΔK147 142-K147 142-ΔK147 142-K147-177 142-ΔK147-177 144-R147-177
rK/09 5,120 640 160 10 640 20 40
rΔK147 1,280 2,560 80–160 10 1,280 20 160
r142-K147 5,120 160 2,560 10 640 20 40
r142-ΔK147 5,120 1,280 320 320 640 320–640 160
r142-K147-177 1,280 80 320 10 320–640 20 10
r142-ΔK147-177 5,120 1,280 160 320 320–640 2,560 10
r144-R147-177 10,240 2,560 640 640 640–1,280 320 640
a
Single values indicate the same HI titers were obtained in three independent experiments. Ranged values indicate that different HI titers were obtained in three independent experiments.

DISCUSSION

It was previously noted that the addition or removal of NLG from the globular head of HA may alter the viral characteristics (39, 40). The introduction of sugar side chains at the tip of HA resulted in the reduced receptor binding ability of the H2 and H5 subtypes (41, 42). However, these two studies presented different perspectives. In the study of H2 HA, two attached N-linked glycans at residues 131, 160, or 197 (H3 numbering; 144, 174, or 211 in H1 numbering, respectively) decreased the polyvalent HA binding avidity to SA, and three NLG sites abolished the fusogenic property of the H2 HA. The study of H5 HA provided somewhat similar results; however, the weak HA-SA binding that was mediated by NLG eventually benefited viral replication by enhancing viral release (43). When considering viral entry to the cell, NLG may decrease viral infectivity by decreasing the strength of the virus-cell interaction. Conversely, when considering the release of viral progeny from the cell, a weak HA-SA bridge may result in the easy release of viral particles, even in cases where the virus retains a neuraminidase (NA) with low enzymatic activity. In our study, mutant viruses retained the same NA protein of wild-type K/09 virus, and no spontaneous mutations in other genes or reverted ones in the HAs were identified even after the multiple passages in eggs, which meant that the altered viral characteristics observed were mainly due to the HA attached N-linked glycans. However, K147-bearing viruses could maintain viral fitness even in the presence of the NLG-driven constraints, whereas ΔK147 viruses experienced reduced viability or lost transmissibility under the same conditions (Fig. 3 and 4).
The biological activity of K147 of H1 HA was recently studied for a structural aspect in relation to interactions between HA and SA (22). Here we suggest a new role of residue 147 as an evolution indicator of H1 HA. In the 1918 H1N1 descendant viruses, residue 147 was occupied by amino acids K (RNA codons AAA or AAG), R (AGA or AGG), or I (AUA), and sometimes it was even deleted (Fig. 1 and Table S1 in the supplemental material). Based on the juxtaposition of residue 147 next to RBS (Fig. 2), we speculated that residue 147 might be inevitably affected by the HA-SA interaction and its resultant polymorphic appearance controlled NLG attachment around the RBS. Indeed, we found that glycosites 144 and 172 were never utilized in the HA-Δ147 hH1N1 viruses (Table 2).
The deletion of residue 147 was intermittently seen in the HA of hH1N1 viruses and then fixed from 1995 to 2009, right before the appearance of 2009 H1N1 viruses. From 1947 to 1986, the HA globular head region of hH1N1 viruses mostly utilized at least three glycosites including 144, 172, and 177 (Table 1). However, no hH1N1 viruses have utilized glycosite 144 since a NLG shift from 144 to 142 in 1986, and glycosite 172 disappeared in 1988 (Table 1). Since then, residues 142 and 177 had been signatures for N-linked glycans in the majority of hH1N1 viruses. This eliminated the need for amino acid 147 required for the glycosites 144 and 172, and the reappearance of the 147 deletion. These findings led us to investigate viral characteristics of mutant viruses having various combinations of NLG patterns within the head of HA and amino acids at residue 147. In our results, r142-ΔK147 and r142-ΔK147-177 viruses exhibited reduced RBC binding ability and replication kinetics in A549 cells (Fig. 3C and E), and they were not transmitted to exposed guinea pigs (Fig. 4G and H). These results indicate that viral fitness is dramatically influenced by N-linked glycans attached at glycosites 142 or 177 in the absence of residue 147. According to a previous study, hH1N1 viruses retaining these kinds of NLG patterns irrespective of residue 147 needed compensation mutation(s) to reduce NLG-driven fitness costs (19).
For HA residue 147, K might be a better genetic signature than R. Although further investigation needs to define the exact effect of R side chain (guanidinium cation) on viral fitness compared to that of other amino acid side chains, the r144-R147-177 virus exhibited reduced RBC binding and replication kinetics in A549 cells in our study (Fig. 3C and E). This virus also less efficiently infected guinea pigs and transmitted to only one out of three exposed guinea pigs (Fig. 4E). Low HI and PRNT titers were then observed in the case of the r144-R147-177 virus-infected guinea pig antiserum (Fig. 5D and Tables 5 and 6). In fact, HA R147-bearing hH1N1 viruses circulating in the past have also appeared to be less immunogenic than HA K147-bearing hH1N1 viruses (44).
Differences in antigenicity were observed according to the presence or absence of K147 in vaccine antigens. For the 1986-1987 influenza season, the World Health Organization (WHO) recommended the A/Chile/1/83 (Ch/83; harboring three glycosylations at 144, 172, and 177) virus as an H1N1 antigen for use in the inactivated trivalent influenza vaccine (TIV) (Table 7) (45). However, since an increasing number of glycosite 142-harboring H1N1 variants were isolated in many geographical regions (46, 47), the WHO had to add a new H1N1 antigen, namely, the A/Singapore/6/86 (Sg/86; glycosylations at 142, 172, and 177) virus, for the 1986–87 influenza season. Until they were replaced with the A/Bayern/7/95 (By/95) virus in 1997, Sg/86-like viruses (including A/Taiwan/1/86 and A/Texas/36/91) represented H1N1 antigens in TIVs (48). One NLG shift from 144 to 142 could effectively cover the Sa antigenic site of Sg/86-like vaccine antigens and thus it may have induced HI reactive antibodies against subsequent hH1N1 variants even though the amino acid variability at the Sa antigenic site increased during the same period (49). In 1998, a new virus, Bj/95, was selected for the H1N1 vaccine antigen. The HA head of the Bj/95 virus held only two NLGs at residues 142 and 177 (Tables 1 and 7). Considering that the Sg/86 vaccine virus controlled many hH1N1 NLG variants during the 1986-1997 influenza seasons and that the difference in NLG patterns between the Sg/86 and Bj/95 viruses seemed to be minor (the Sg/86 virus had three NLGs at residues 142, 172, and 177 within the HA head region, and the Bj/95 virus had only two NLGs at residues 142 and 177; the amino acid difference between two viruses is calculated only as 5.81% within HA1 sequences), there might be another reason for the antigenic difference between the Sg/86 and Bj/95 viruses. Thus, it appears that the absence of K147 in Bj/95 may have contributed to antigenic differences. In the HI results derived from the use of postinfection ferret antisera, Bj/95 antiserum reacted only with the HA-ΔK147viruses and were less reactive to Sg/86- and By/95-like viruses, which retained K147 in their HAs (50, 51). A similar result was also seen in our HI assays. The r142-177 antiserum induced more than 640 HI titers to homologous or heterologous viruses and even produced 80 HI titers to the r142-ΔK147-177 virus (Table 5). However, the r142-ΔK147-177 antiserum resulted in only 10 to 40 HI titers to the HA-K147-bearing viruses (Table 5). Considered together, the selected H1N1 vaccine antigens reflect how the HA of hH1N1 viruses has evolved against human immune responses and the genetic signature of HA residue 147 has likely impacted the immunogenicity of the selected H1N1 vaccine antigens.
Table 7
Table 7 hH1N1 vaccine viruses and the NLG status and amino acids of residue 147 at the globular head of HA
Yr Vaccine virus GenBank no. Residues harboring N-linked glycans and amino acids from residue 147 at the globular head of HAa
142 144 147 172 177
1982–83 A/Brazil/11/78 ABO38065   o R o o
1984–86 A/Chile/1/83 ABO38340   o K o o
1987–96 A/Singapore/6/86 ABO38395 o   K o o
  A/Taiwan/1/86 ABF21274 o   K o  
  A/Texas/36/91 AAP34322 o   K   o
1997 A/Bayern/7/95 CAD29944 o   K   o
1998–99 A/Beijing/262/95 ACF41867 o   Absent   o
2000–06 A/New Caledonia/20/99 ACF41878 o   Absent   o
2007 A/Solomon Islands/3/2006 ABU50586 o   Absent   o
2008–09 A/Brisbane/59/2007 AET50439 o   Absent   o
2010–12 A/California/7/2009 AFM72832     K    
a
See Table 1, footnote a.
Over the past several decades of circulation, hH1N1 viruses have responded against external pressures, such as neutralizing antibodies, and the HA of hH1N1 viruses has evolved through changing antigenicity. Similar to the evolution pathway of the 1918 virus, some descendants of the 2009 H1N1 virus already utilized HA glycosite 179 (Table 8) (12) and selected arginine, instead of lysine, as a genetic signature for HA residue 147 (Table 9). Even though it is uncertain how future hH1N1 viruses may or not recapitulate the evolutionary pathway of the old H1N1s, we suggest that the genetic signature at HA residue 147 is an indicator for the evolution dynamics of hH1N1 viruses and that the presence of this amino acid allows for a broader possible use of additional NLGs within the globular head region of HA. Also, the presence of amino acid 147 increases the breadth of the antibody response induced by natural infection.
Table 8
Table 8 Reported 2009 H1N1 variants retaining the HA S179N mutation (after identical sequences were collapsed)
Virus GenBank no. HA amino acid residues at selected positions
142 144 147 172 177 179 181
A/Korea/01/2009 (S179 control) ACQ84451 N D K G K S S
A/Scotland/98/2009 AEO01518 N D K G K N S
A/Wurzburg/INS381/2009 ADM13031 N D K G K N S
A/District of Columbia/WRAIR0310/2010 AEM92378 N D K G K N S
A/Ecuador/IEC00012/2010 AER92719 N D K G K N S
A/Ecuador/IEC00027/2010 AER92720 N D K G K N S
A/Ecuador/IEC00032/2010 AER92722 N D K G K N S
A/Ecuador/IEC00043/2010 AER92723 N D K G K N S
A/Mexico City/WRAIR3569N/2010 AEM92528 N D K G K N S
A/Tallin/INS183/2010 ADG42553 N D K G K N S
A/Tallin/INS374/2010 ADM31858 N D K G K N S
A/Warsaw/INS316/2010 ADM31588 N D K G K N S
A/Mexico/InDRE1945/2011 AEA74031 N D K G K N S
A/Shenzhen/lg56/2011 AEV40218 N D K G K N S
A/Shenzhen/ns06/2011 AEV40220 N D K G K N S
A/South Carolina/NHRC0002/2011 AEJ10500 N D K G K N S
A/Tianjinhedong/SWL44/2011 AFE11257 N D K G K N S
A/Tianjinjinnan/SWL41/2011 AFE11247 N D K G K N S
A/Wisconsin/28/2011 AEX38474 N D R G K N S
A/Ontario/N163578/2012 AFV31495 N D R G K N S
Table 9
Table 9 Reported 2009 H1N1 variants retaining the HA K147R mutation (after identical sequences were collapsed)
Virus GenBank no. HA amino acid residues at selected positions
142 144 147 172 177 179 181
A/Korea/01/2009 (K147 control) ACQ84451 N D K G K S S
A/Jiangsu/ALS1/2011 ADW01407 N D R G K S S
A/Wisconsin/28/2011 AEX38474 N D R G K N S
A/Ontario/N163578/2012 AFV31495 N D R G K N S

ACKNOWLEDGMENTS

This study was supported by grants from the Korea Healthcare Technology R&D Project of the Ministry of Health and Welfare (grant A103001), the Korea Centers for Diseases Control and Prevention (grant 2010-E43002-00), and the Hallym University Specialization Fund (HRF-S-41). This study was also partly supported by the Center for Research on Influenza Pathogenesis, a National Institute of Allergy and Infectious Diseases-funded Center of Excellence in Influenza Research and Surveillance (HHSN266200700010C) (to A.G.-S.).
We thank Peter Palese (Department of Microbiology, Mount Sinai School of Medicine, New York, NY) for providing plasmids for the reverse genetics system. We also thank Saem Shin and Sulhwa Jung for technical assistance with the animal experiments.

Supplemental Material

File (zjv999097793so1.pdf)
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
World Health Organization. 2012. Fact sheet on influenza. World Health Organization, Geneva, Switzerland. http://www.who.int/mediacentre/factsheets/fs211/en/index.html.
2.
Palese P. 2004. Influenza: old and new threats. Nat. Med. 10:S82–S87.
3.
Smith GJ, Vijaykrishna D, Bahl J, Lycett SJ, Worobey M, Pybus OG, Ma SK, Cheung CL, Raghwani J, Bhatt S, Peiris JS, Guan Y, and Rambaut A. 2009. Origins and evolutionary genomics of the 2009 swine-origin H1N1 influenza A epidemic. Nature 459:1122–1125.
4.
Neumann G and Kawaoka Y. 2011. The first influenza pandemic of the new millennium. Influenza Other Respir. Viruses 5:157–166.
5.
Hancock K, Veguilla V, Lu X, Zhong W, Butler EN, Sun H, Liu F, Dong L, DeVos JR, Gargiullo PM, Brammer TL, Cox NJ, Tumpey TM, and Katz JM. 2009. Cross-reactive antibody responses to the 2009 pandemic H1N1 influenza virus. N. Engl. J. Med. 361:1945–1952.
6.
Itoh Y, Shinya K, Kiso M, Watanabe T, Sakoda Y, Hatta M, Muramoto Y, Tamura D, Sakai-Tagawa Y, Noda T, Sakabe S, Imai M, Hatta Y, Watanabe S, Li C, Yamada S, Fujii K, Murakami S, Imai H, Kakugawa S, Ito M, Takano R, Iwatsuki-Horimoto K, Shimojima M, Horimoto T, Goto H, Takahashi K, Makino A, Ishigaki H, Nakayama M, Okamatsu M, Warshauer D, Shult PA, Saito R, Suzuki H, Furuta Y, Yamashita M, Mitamura K, Nakano K, Nakamura M, Brockman-Schneider R, Mitamura H, Yamazaki M, Sugaya N, Suresh M, Ozawa M, Neumann G, Gern J, Kida H, Ogasawara K, and Kawaoka Y. 2009. In vitro and in vivo characterization of new swine-origin H1N1 influenza viruses. Nature 460:1021–1025.
7.
2009. Serum cross-reactive antibody response to a novel influenza A (H1N1) virus after vaccination with seasonal influenza vaccine. MMWR Morb. Mortal. Wkly. Rep. 58:521–524.
8.
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, Donis R, Katz J, Finelli L, Bridges CB, Shaw M, Jernigan DB, Uyeki TM, Smith DJ, Klimov AI, and Cox NJ. 2009. Antigenic and genetic characteristics of swine-origin 2009 A(H1N1) influenza viruses circulating in humans. Science 325:197–201.
9.
Xu R, Ekiert DC, Krause JC, Hai R, Crowe JE Jr, and Wilson IA. 2010. Structural basis of preexisting immunity to the 2009 H1N1 pandemic influenza virus. Science 328:357–360.
10.
Stevens J, Corper AL, Basler CF, Taubenberger JK, Palese P, and Wilson IA. 2004. Structure of the uncleaved human H1 hemagglutinin from the extinct 1918 influenza virus. Science 303:1866–1870.
11.
Shope RE. 1936. The incidence of neutralizing antibodies for swine influenza virus in the sera of human beings of different ages. J. Exp. Med. 63:669–684.
12.
Wei CJ, Boyington JC, Dai K, Houser KV, Pearce MB, Kong WP, Yang ZY, Tumpey TM, and Nabel GJ. 2010. Cross-neutralization of 1918 and 2009 influenza viruses: role of glycans in viral evolution and vaccine design. Sci. Transl. Med. 2:24ra21.
13.
Cohen J. 2010. Swine flu pandemic. What's old is new: 1918 virus matches 2009 H1N1 strain. Science 327:1563–1564.
14.
Manicassamy B, Medina RA, Hai R, Tsibane T, Stertz S, Nistal-Villan E, Palese P, Basler CF, and Garcia-Sastre A. 2010. Protection of mice against lethal challenge with 2009 H1N1 influenza A virus by 1918-like and classical swine H1N1 based vaccines. PLoS Pathog. 6:e1000745.
15.
Gerhard W and Webster RG. 1978. Antigenic drift in influenza A viruses. I. Selection and characterization of antigenic variants of A/PR/8/34 (HON1) influenza virus with monoclonal antibodies. J. Exp. Med. 148:383–392.
16.
Skehel JJ, Stevens DJ, Daniels RS, Douglas AR, Knossow M, Wilson IA, and Wiley DC. 1984. A carbohydrate side chain on hemagglutinins of Hong Kong influenza viruses inhibits recognition by a monoclonal antibody. Proc. Natl. Acad. Sci. U. S. A. 81:1779–1783.
17.
Schulze IT. 1997. Effects of glycosylation on the properties and functions of influenza virus hemagglutinin. J. Infect. Dis. 176(Suppl 1):S24–S28.
18.
Kim JI and Park MS. 2012. N-linked glycosylation in the hemagglutinin of influenza A viruses. Yonsei Med. J. 53:886–893.
19.
Das SR, Hensley SE, David A, Schmidt L, Gibbs JS, Puigbo P, Ince WL, Bennink JR, and Yewdell JW. 2011. Fitness costs limit influenza A virus hemagglutinin glycosylation as an immune evasion strategy. Proc. Natl. Acad. Sci. U. S. A. 108:E1417–1422.
20.
Das SR, Puigbo P, Hensley SE, Hurt DE, Bennink JR, and Yewdell JW. 2010. Glycosylation focuses sequence variation in the influenza A virus H1 hemagglutinin globular domain. PLoS Pathog. 6:e1001211.
21.
Sun S, Wang Q, Zhao F, Chen W, and Li Z. 2011. Glycosylation site alteration in the evolution of influenza A (H1N1) viruses. PLoS One 6:e22844.
22.
Xu R, McBride R, Nycholat CM, Paulson JC, and Wilson IA. 2012. Structural characterization of the hemagglutinin receptor specificity from the 2009 H1N1 influenza pandemic. J. Virol. 86:982–990.
23.
Reed LJ and Muench H. 1938. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27:493–497.
24.
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.
25.
Hensley SE, Das SR, Bailey AL, Schmidt LM, Hickman HD, Jayaraman A, Viswanathan K, Raman R, Sasisekharan R, Bennink JR, and Yewdell JW. 2009. Hemagglutinin receptor binding avidity drives influenza A virus antigenic drift. Science 326:734–736.
26.
Gamblin SJ, Haire LF, Russell RJ, Stevens DJ, Xiao B, Ha Y, Vasisht N, Steinhauer DA, Daniels RS, Elliot A, Wiley DC, and Skehel JJ. 2004. The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science 303:1838–1842.
27.
Soundararajan V, Tharakaraman K, Raman R, Raguram S, Shriver Z, Sasisekharan V, and Sasisekharan R. 2009. Extrapolating from sequence–the 2009 H1N1 ‘swine’ influenza virus. Nat. Biotechnol. 27:510–513.
28.
Kim JI, Lee I, Park S, and Park M-S. 2012. Surface glycoproteins determine the feature of the 2009 pandemic H1N1 virus. BMB Rep. 45:653–658.
29.
Medeiros R, Naffakh N, Manuguerra JC, and van der Werf S. 2004. Binding of the hemagglutinin from human or equine influenza H3 viruses to the receptor is altered by substitutions at residue 193. Arch. Virol. 149:1663–1671.
30.
Maines TR, Chen LM, Van Hoeven N, Tumpey TM, Blixt O, Belser JA, Gustin KM, Pearce MB, Pappas C, Stevens J, Cox NJ, Paulson JC, Raman R, Sasisekharan R, Katz JM, and Donis RO. 2011. Effect of receptor binding domain mutations on receptor binding and transmissibility of avian influenza H5N1 viruses. Virology 413:139–147.
31.
Scott JN, Nucci NV, and Vanderkooi JM. 2008. Changes in water structure induced by the guanidinium cation and implications for protein denaturation. J. Phys. Chem. A 112:10939–10948.
32.
Kilbourne ED. 1977. Influenza pandemics in perspective. JAMA 237:1225–1228.
33.
Reperant LA, Kuiken T, and Osterhaus AD. 2012. Adaptive pathways of zoonotic influenza viruses: from exposure to establishment in humans. Vaccine 30:4419–4434.
34.
Ito T and Kawaoka Y. 2000. Host-range barrier of influenza A viruses. Vet. Microbiol. 74:71–75.
35.
Belser JA, Maines TR, Tumpey TM, and Katz JM. 2010. Influenza A virus transmission: contributing factors and clinical implications. Expert Rev. Mol. Med. 12:e39.
36.
Thompson CI, Barclay WS, Zambon MC, and Pickles RJ. 2006. Infection of human airway epithelium by human and avian strains of influenza A virus. J. Virol. 80:8060–8068.
37.
Skehel JJ and Wiley DC. 2000. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu. Rev. Biochem. 69:531–569.
38.
Lowen AC, Mubareka S, Tumpey TM, Garcia-Sastre A, and Palese P. 2006. The guinea pig as a transmission model for human influenza viruses. Proc. Natl. Acad. Sci. U. S. A. 103:9988–9992.
39.
Deshpande KL, Fried VA, Ando M, and Webster RG. 1987. Glycosylation affects cleavage of an H5N2 influenza virus hemagglutinin and regulates virulence. Proc. Natl. Acad. Sci. U. S. A. 84:36–40.
40.
Roberts PC, Garten W, and Klenk HD. 1993. Role of conserved glycosylation sites in maturation and transport of influenza A virus hemagglutinin. J. Virol. 67:3048–3060.
41.
Tsuchiya E, Sugawara K, Hongo S, Matsuzaki Y, Muraki Y, Li ZN, and Nakamura K. 2002. Effect of addition of new oligosaccharide chains to the globular head of influenza A/H2N2 virus haemagglutinin on the intracellular transport and biological activities of the molecule. J. Gen. Virol. 83:1137–1146.
42.
Ohuchi M, Ohuchi R, Feldmann A, and Klenk HD. 1997. Regulation of receptor binding affinity of influenza virus hemagglutinin by its carbohydrate moiety. J. Virol. 71:8377–8384.
43.
Wagner R, Wolff T, Herwig A, Pleschka S, and Klenk HD. 2000. Interdependence of hemagglutinin glycosylation and neuraminidase as regulators of influenza virus growth: a study by reverse genetics. J. Virol. 74:6316–6323.
44.
Daniels RS, Douglas AR, Skehel JJ, and Wiley DC. 1985. Antigenic and amino acid sequence analyses of influenza viruses of the H1N1 subtype isolated between 1982 and 1984. Bull. World Health Organ. 63:273–277.
45.
World Health Organization. 1986. Recommended composition of influenza virus vaccines for use in the 1986-1987 season. Wkly. Epidemiol. Rec. 61:61–64.
46.
World Health Organization. 1986. Composition of influenza virus vaccines for use in the 1986-1987 season: an update. Wkly. Epidemiol. Rec. 61:237–244.
47.
Centers for Disease Control and Prevention. 1986. Monovalent influenza A(H1N1) vaccine, 1986-1987. MMWR Morb. Mortal. Wkly. Rep. 35:517–521.
48.
World Health Organization. 1996. WHO influenza surveillance. Wkly. Epidemiol. Rec. 71:353–357.
49.
Sun S, Wang Q, Zhao F, Chen W, and Li Z. 2012. Prediction of biological functions on glycosylation site migrations in human influenza H1N1 viruses. PLoS One 7:e32119.
50.
World Health Organization. 1997. Recommended composition of influenza virus vaccines for use in the 1997-1998 season. Wkly. Epidemiol. Rec. 72:57–61.
51.
World Health Organization. 1998. Recommended composition of influenza virus vaccines for use in the 1998-1999 season. Wkly. Epidemiol. Rec. 73:56–61.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 87Number 131 July 2013
Pages: 7539 - 7549
PubMed: 23637398

History

Received: 5 February 2013
Accepted: 20 April 2013
Published online: 1 July 2013

Permissions

Request permissions for this article.

Contributors

Authors

Jin Il Kim
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Ilseob Lee
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Sehee Park
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Min-Woong Hwang
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Joon-Yong Bae
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Sangmoo Lee
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Jun Heo
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Mee Sook Park
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Adolfo García-Sastre
Department of Microbiology, Mount Sinai School of Medicine, New York, New York, USA
Department of Medicine, Division of Infectious Diseases, Mount Sinai School of Medicine, New York, New York, USA
Global Health and Emerging Pathogens Institute, Mount Sinai School of Medicine, New York, New York, USA
Man-Seong Park
Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea
Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Korea

Notes

Address correspondence to Man-Seong Park, [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