Abstract
Frequent interspecies transmission of human influenza A viruses (FLUAV) to pigs contrasts with the limited subset that establishes in swine. While hemagglutinin mutations are recognized for their role in cross-species transmission, the contribution of neuraminidase remains understudied. Here, the NA’s role in FLUAV adaptation was investigated using a swine-adapted H3N2 reassortant virus with human-derived HA and NA segments. Adaptation in pigs resulted in mutations in both HA (A138S) and NA (D113A). The D113A mutation abolished calcium (Ca2+) binding in the low-affinity Ca2+-binding pocket of NA, enhancing enzymatic activity and thermostability under Ca2+-depleted conditions, mirroring swine-origin FLUAV NA behavior. Structural analysis predicts that swine-adapted H3N2 viruses lack Ca2+ binding in this pocket. Further, residue 93 in NA (G93 in human, N93 in swine) also influences Ca2+ binding and impacts NA activity and thermostability, even when D113 is present. These findings demonstrate that mutations in influenza A virus surface proteins alter evolutionary trajectories following interspecies transmission and reveal distinct mechanisms modulating NA activity during FLUAV adaptation, highlighting the importance of Ca2+ binding in the low-affinity calcium-binding pocket.
Similar content being viewed by others
Introduction
Interspecies transmission of Influenza A viruses (FLUAV) is common; however, sustained transmission within a new host species is rare. This is because the virus must undergo adaptive evolution to replicate, transmit, and become endemic in the new host population. FLUAV have two major surface glycoproteins: hemagglutinin (HA) and neuraminidase (NA). The HA recognizes sialic acids (SAs) on the cell surface and triggers virus entry into the host cells1,2. The NA shows an opposite activity, cleaving SAs to allow the virus to move through the respiratory mucus by preventing binding to decoy receptors and to promote virus release from infected cell3,4.
Due to its central role in the infection process, the HA protein is considered the primary driver of adaptation to a new host species. This adaptation often involves the acquisition of mutations in the receptor binding site (RBS) that enhance affinity for host-specific SA conformations5,6,7. Previous studies have demonstrated that adaptation of FLUAVs isolated from humans to pigs results in amino acid changes in the HA RBS, increasing affinity for swine airway epithelial cells8 and improving transmission efficiency compared to the original human virus9. However, other gene segments also contribute to the adaptation of human-origin FLUAVs to pigs, with the acquisition of a well-adapted internal gene segment constellation seemingly essential10,11,12. Human FLUAVs introduced to pigs frequently lose their internal gene segments through reassortment, acquiring swine-adapted gene segments from endemic strains13. Interestingly, the surface gene segments (HA and NA) often persist in the swine population, albeit with significant genetic differences from the precursor strains13,14, leading to the establishment of new lineages.
The NA protein is a critical factor in FLUAV adaptation across species, as it must maintain a functional balance with HA for successful infection15. Adaptive mutations in the HA RBS can disrupt this balance, often prompting compensatory mutations in the NA protein16. NA is a homotetrameric enzyme with hydrolase activity, with each monomer consisting of a transmembrane domain, a stalk domain, and a head domain containing the catalytic pocket17,18,19. While the catalytic pocket is present in each monomer, optimal sialidase activity is only achieved in the tetrameric state20,21,22. Calcium (Ca2+) is an essential cofactor for NA activity, and most NA subtypes possess a high-affinity Ca2+-binding pocket near the active site of each monomer23,24,25. This binding event is thought to induce a conformational change in the catalytic site, facilitating proper SA binding26,27. In addition to the high-affinity site, certain subtypes such as N1 and N2 NAs have a second, low-affinity Ca2+-binding pocket located within the symmetry axis of the tetramer27,28. This second site is believed to contribute to tetramer stability and, indirectly, to NA activity, as NA activity is dependent on the tetrameric form28,29,30. Notably, the crystal structure of the 2009 pandemic H1N1 virus NA revealed that the virus quickly changed its calcium preference in the symmetry axis after jumping from pigs to humans28.
Despite these observations, the precise role of the second Ca2+-binding site in the adaptation of FLUAV between species remains poorly understood and requires further investigation. In this study, we investigated the adaptive mechanisms of human-derived FLUAV HA and NA gene segments during transmission in a swine host. Utilizing a reassortant virus carrying HA and NA segments from the human strain A/Victoria/361/2011 (hVIC/11) in a swine-origin FLUAV backbone. The rapid emergence of an adaptative mutation was observed in HA (A138S, H3 numbering—herein referred to as the hVIC/11-A138S strain) near the RBS that evolved after infection of pigs8. Subsequently, and after two serial transmission events in vivo in pigs, the emergence and fixation of a novel mutation in the NA, D113A, was observed. In silico structural analyses revealed that amino acid 113 in the N2 NA of hVIC/11 interacts with Ca2+ in the low-affinity Ca2+-binding pocket, a characteristic typically observed in human H3N2 strains. Remarkably, the D113A mutation abolished Ca2+ binding in this pocket, but concurrently enhanced NA thermostability and activity under Ca2+-depleted conditions. However, the A113 variant is uncommon in both human and swine H3N2 strains, and, notably, swine-origin H3N2 FLUAVs harboring this mutation are predicted to be unable to bind Ca2+ in the low-affinity pocket. This observation prompted further investigation into the molecular determinants of this differential binding behavior, which revealed an important role of amino acid at position 93 in NA that further influences Ca2+ binding in the low-affinity pocket and impacts replication and transmission efficiency in pigs. These findings highlight the intricate molecular determinants of FLUAV adaptation between hosts and underscore the critical role of Ca2+ binding in this process.
Results
hVIC/11-A138S infects the lower respiratory tract and increases affinity following transmission to contact animals
To evaluate adaptation to a new host, 3-week-old pigs (seeders) were inoculated intratracheally and intranasally with 3 × 106 TCID50/pig of either hVIC/11-A138S9 or the control swine-adapted virus sOH/0431. Two days post-inoculation (dpi), three naïve pigs (contact 1, C1) were introduced with the inoculated pigs (seeders, Fig. 1). At 5 dpi, seeders were euthanized, and three new naïve pigs (contact 2, C2) were introduced to the C1 pigs. This cycle was repeated for a total of four contact groups. Nasal swabs were collected throughout the study, and lung samples and bronchoalveolar lavage fluid (BALF) were collected during necropsies at 5 dpi.
Pigs were challenged with either sOH/04 or hVIC/11-A138S and nasal swabs were collected. Lung tissue and BALF samples were collected during necropsies at 5 dpi/6 dpc. a Viral titers in nasal swab samples (n = 3 per contact) from pigs challenged with hVIC/11-A138S. b Viral titers in nasal swab samples (n = 3 per contact) from pigs challenged with sOH/04. Viral titers from the trachea (c), right cranial lobe (d), left cranial lobe (e), right caudal lobe (f), left caudal lobe (g), bronchoalveolar lavage fluid (BALF) (h), and the accessory lobe (i) from hVIC/11-A138S-infected pigs were determined by RT-qPCR and normalizing all samples to 1 μg total RNA. All statistical analyses were performed by two-way ANOVA. Values represent the mean ± SD.
Virus titration of nasal swabs confirmed successful transmission of hVIC/11-A138S among all contacts, peaking at approximately 105 TCID50eq/mL by 3 dpi (Fig. 1a). The sOH/04 virus control was also detected in all contacts (Fig. 1b), albeit at higher titers (approximately 106 TCID50eq/mL). The hVIC/11-A138S virus infected the trachea of all seeders and contact pigs (Fig. 1c). Virus titers in the right cranial, left cranial, and right caudal lobes (Fig. 1d–f) increased significantly by contact 4 (approximately 104 TCID50eq/mL) compared to the seeder and/or C1 pigs. While no significant differences were observed in the left caudal lobe or BALF samples (Fig. 1g, h), although a trend towards higher titers was noted in contact 4 pigs’ BALF samples. Notably, the virus initially failed to infect the accessory lobe of seeders and C1 pigs (Fig. 1i), with titers below the limit of detection (<101 TCID50eq/mL) but was detected in subsequent contacts from C2 onward (approximately 103 TCID50eq/mL). These findings suggest that hVIC/11-A138S replication efficiency in the lower respiratory tract increased with each transmission, ultimately infecting all lung lobes by the study’s conclusion.
Serial transmission of hVIC/11-A138S in pigs selected for a mutant with a single amino acid mutation in NA
After sequencing analysis of nasal swabs and BALF samples, only minor variants (present in less than 50% of sequences) were observed in the hVIC/11-A138S HA segment (Fig. 2a), and none of these variants became fixed or were transmitted. However, a mutation in the NA, D113A, was detected in contact 2 pigs as a major variant (present in more than 50% of sequences) that eventually became prevalent and was transmitted to all contact 3 and 4 pigs (Fig. 2b). No positive selection was observed in the HA segment (Supplementary Fig. 1a), while signs of positive selection were observed for the NA segment in contacts 1 and 2 sequences (Supplementary Fig. 1b). After acquisition of the D113A amino acid change, no positive selection was observed in this segment. No major changes were observed in the sOH/04 control virus surface gene segments (Supplementary Fig. 2). Using the NA crystal structure information that closely matches the sequence of hVIC/11 NA (A/Tanzania/205/2010, Supplementary Fig. 3) revealed that the amino acid at position 113 located within the low-affinity Ca2+-binding pocket of the NA is essential for Ca2+ binding (Fig. 2c). The polar side chain of aspartic acid (D) at this position is predicted to stabilize a water molecule via a hydrogen bond (one per monomer) in the symmetry axis (Fig. 2c, top panel), facilitating the coordination of a single Ca2+ ion. When D113 is mutated to alanine (A), this polar interaction is lost, likely impairing Ca2+ binding (Fig. 2c, bottom panel). Notably, temporal analysis of residue frequency reveals that D113 and A113 were similarly prevalent in swine H3N2 viruses until the early 2000s (Fig. 2d). However, D113 has since become predominant, reaching 100% frequency by 2022.
Nasal swab samples collected at days 1, 2, and 5 dpi and 1, 3, and 6 dpc were sequenced and viral variants across the HA (a) and NA (b) segments were identified and their abundance was quantified. Mutations were classified as nonsynonymous (yellow) or synonymous (blue). A minimum threshold for major variants was arbitrarily set at 0.5 and the fixation point at a frequency of 1 (dashed lines). The frequency is expressed in the y axis in a log scale from 0 to 1. The data represents the diversity observed in three pigs per contact. (c) hVIC/11 NA tetramer protein model showing the low-affinity Ca2+ -binding pocket when D113 and A113 are present. Residue 113 is shown in blue, water molecules in red, and the Ca2+ atom in purple. Images were made using PyMOL. d Frequency of A113 in swine H3N2 isolated reported from 1992 to 2023. Sequences were obtained from GISAID and aligned with Clustal Omega. Alanine (A) is shown in green while aspartic acid (D) is presented in red.
The adaptive N2 D113A mutation enhances NA stability under Ca2+- depleted conditions and reduces Ca2+ requirements for NA sialidase activity
Prior research with the N1 NA from the 2009 pandemic H1N1 virus demonstrated that disrupting Ca2+ binding in the low-affinity Ca2+-binding pocket impairs virus replication in vitro28. To investigate this effect in viruses with the N2 NA D113A mutation, two recombinant viruses were produced carrying this mutation, with or without the HA A138S mutation (hVIC/11-A138S/D113A and hVIC/11-D113A, respectively). Replication kinetics of these viruses were evaluated in various human and swine cell lines (Fig. 3a) and compared to hVIC/11-A138S, hVIC/11, and sOH/04 viruses. The N2 NA D113A mutation did not affect virus replication in MDCK, PK15, A549, or differentiated human airway epithelial (HAE) cells (Fig. 3a). Neither hVIC/11 nor hVIC/11-D113A replicated in differentiated primary swine airway epithelial (SAE) cells, unlike hVIC/11-A138S and hVIC/11-A138S/D113A, confirming the advantage conferred by the HA A138S mutation for replication in swine cells9.
a Viral growth kinetics of sOH/04 (blue), hVIC/11 (yellow), hVIC/11-A138S(red), hVIC/11-D113A (black), and hVIC/11-A138S/D113A (purple) in MDCK, PK15, A549, HAE, and SAE cells infected at an MOI of 0.01. A mock control (white) is included. Values represent the mean ± SD of three independent experiments. p values were obtained by ordinary one-way ANOVA. b Thermostability of sOH/04 (blue), hVIC/11 (yellow), hVIC/11-A138S(red), hVIC/11-D113A (black), hVIC/11-A138S/D113A (purple) NAs determined in presence (solid line) or absence (dashed line) of Ca2+. c T50 was obtained by adjusting the data to a dose-response variable slope nonlinear fit and values are shown as the means of three independent experiments. p values were obtained by ordinary one-way ANOVA. d NA sialidase activity of sOH/04 (blue), hVIC/11 (yellow), hVIC/11-A138S(red), hVIC/11-D113A (black), and hVIC/11-A138S/D113A (purple) at 0 mM Ca2+ expressed as a percentage of the reaction velocity compared to 2 mM Ca2+ (100% NA activity) at 37 °C. Values represent the means ± SD of three independent experiments. p values were obtained by ordinary one-way ANOVA.
Disruption of Ca2+ binding in the symmetry axis may negatively impact NA stability28. To investigate this, the temperature at which 50% of NA activity is lost (T50) was established for NAs containing either D113 or A113 (Fig. 3b) in the absence or in the presence of physiological Ca2+ concentration (2 mM). The assay revealed that introducing the D113A mutation decreased the T50 of NA proteins at 2 mM Ca2+ (hVIC/11-D113A, 52.03 °C; hVIC/11-A138S/D113A, 51.19 °C) compared to control viruses with wild-type (WT) NA (hVIC/11, hVIC/11-A138S, and sOH/04), which exhibited T50 values averaging 5 °C higher (Fig. 3b, left panel; Fig. 3c). In the absence of Ca2+, all T50 values decreased compared to those at 2 mM Ca2+ (Fig. 3b, right panel). Remarkably, viruses carrying D113A, particularly hVIC/11-A138S/D113A (T50 44.44 °C), showed significantly higher values than the WT hVIC/11 (T50 39.51 °C) in the absence of Ca2+, behaving more like the sOH/04 strain (T50 43.40 °C) (Fig. 3c). These findings suggest that disrupting Ca2+ binding in the symmetry axis decreases NA thermostability at 2 mM Ca2+ but stabilizes the protein in the absence of Ca2+.
The sialidase activity in the absence of Ca2+as a percentage of the activity at 2 mM Ca2+ measured at 37 °C (Fig. 3d, Supplementary Fig. 4) revealed that mutant viruses containing the A113 mutation (hVIC/11-A138S/D113A and hVIC/11-D113A) maintained high levels of NA activity under Ca2+-depleted conditions similar to sOH/04 (39.14%). These mutants also exhibited enhanced activity compared to hVIC/11, which was nearly inactivated in the absence of Ca2+ (3.81%). Notably, the virus with both the A138S mutation in the HA and the D113A mutation in the NA retained the highest levels of sialidase activity in the absence of Ca2+, exceeding 50%.
Ca2+ binding within the low-affinity Ca2+ binding pocket is influenced by amino acid 93
Analysis of the predicted hVIC/11 and sOH/04 and NA structures (sOH/04 model derived from homology with A/Moscow/10/1999 (H3N2), Supplementary Fig. 5), suggests that the latter likely does not bind Ca2+ in this pocket despite having D113 (Fig. 4a). This model indicates altered interactions between sOH/04 NA and water molecules within the symmetry axis compared to hVIC/11, disrupting Ca2+ coordination. This is attributed to a reduced ϕ angle between G111 and G112 in sOH/04’s NA (159.3°) relative to hVIC/11’s NA (176.8°), which repositions the D113 side chain (Supplementary Fig. 6a). To verify this, the D113A mutation was introduced into sOH/04 NA (sOH/04-D113A). This did not alter NA activity at varying Ca2+ concentrations compared to WT sOH/04 NA (Fig. 4b) nor affected thermal stability at 2 mM or 0 mM Ca2+, further supporting the notion that sOH/04 NA does not bind Ca2+ in this pocket (Fig. 4c). Sequence alignment revealed an important difference at position 93, in which asparagine (N) is present in sOH/04 NA and glycine (G) in hVIC/11 NA (Supplementary Fig. 6b). Temporal analysis of human and swine H3N2 NA sequences (1992-2023) showed residue 93 in human isolates shifting from K (lysine) to N to D by the early 2010s, with G becoming prevalent as of 2023 (Supplementary Fig. 6c and d). Swine H3N2 viruses initially carried K93, gradually replaced by N93 (Supplementary Fig. 6c, bottom panel), which became fixed by the 2010s and has not changed ever since. This change at position 93 appears to affect subsequent residues, potentially explaining the altered ϕ angle between G111 and G112 (Supplementary Fig. 6e). This is further supported by comparing NA activity of swine- and human-origin H3N2 FLUAVs at different Ca2+ concentrations. Human H3N2 FLUAVs have exhibited reduced tolerance to Ca2+ depletion since the early 2000s (Supplementary Fig. 7a–i), contrasting with swine H3N2 strains, which largely retain high sialidase activity in the absence of Ca2+ (Supplementary Fig. 7j–q). This effect on NA activity seems to be associated with the residue at position 93 (Supplementary Table S1).
a NA protein model showing the interaction of G111, G112, and D113 with water molecules in the symmetry axis of the NA tetramer of hVIC/11 (right panel, green) and sOH/04 (left panel, blue). Water molecules are shown in red, and Ca2+ is shown in green. b sOH/04 (blue) and sOH/04-D113A (pink) NA sialidase activity at different Ca2+ concentrations. Values are presented as a percentage of the reaction velocity compared to 2 mM Ca2+ which was set at 100%, measured at 37 °C. Values represent the mean ± SD of three independent experiments. c sOH/04 and sOH/04-D113A thermostability assessed at 2 mM Ca2+ (solid line) or 0 mM Ca2+ (dashed line). T50 was determined by adjusting the data of three independent experiments to a dose-response variable slope nonlinear fit. d Left panel: Thermostability of hVIC/11 (yellow), hVIC/11-G93N (brown), and hVIC/11-A138S/G93N (green). Right panel: Thermostability of sOH/04 (blue) and sOH/04-N93G (orange). Thermostability profiles were determined at 2 mM Ca2+ (solid line) or 0 mM Ca2+ (dashed line). Values represent the mean ± SD of three independent experiments. e T50 values were determined by adjusting the data of three independent experiments to a dose-response variable slope nonlinear fit. p values were obtained by ordinary one-way ANOVA. f Left panel: NA sialidase activity of hVIC/11 (yellow), hVIC/11-G93N (brown), and hVIC/11-A138S/G93N (green) at different Ca2+ concentrations. Right panel: NA sialidase activity of sOH/04 (blue) and sOH/04-N93G (orange) at different Ca2+ concentrations. Values are expressed as the reaction velocity percentage compared to the velocity at 2 mM Ca2+ (100% activity), measured at 37 °C. Values represent the mean ± SD of three independent experiments. (g) NA activity at 0 mM Ca2+ of hVIC/11 and sOH/04 NA 93 mutants expressed as a percentage of the reaction velocity compared to 2 mM Ca2+ (100% NA activity), measured at 37 °C. Values are shown as the means of three independent experiments and p values were obtained by ordinary one-way ANOVA.
To investigate the influence of residue 93 on NA physiochemical properties, the N93 substitution was introduced into the hVIC/11 NA, generating both hVIC/11-G93N and hVIC/11-A138S/G93N mutants. While the presence of Ca2+ led to a slight decrease in T50 compared to the unmodified hVIC/11, the absence of Ca2+ revealed a distinct pattern. In this Ca2+-depleted environment, the hVIC/11-G93N mutant exhibited a significant increase in T50 (47.22 °C) compared to both hVIC/11 and hVIC/11-A138S/G93N (Fig. 4d, e). The impact of residue 93 was not limited to the hVIC/11 NA. The N93G mutation in sOH/04-N93G NA also affected stability, resulting in reduced T50 values under both Ca2+-replete and Ca2+-depleted conditions compared to the sOH/04 control virus. These observations underscore the consistent influence of residue 93 on NA stability, regardless of the specific NA mutant or Ca2+ availability. Interestingly, this influence on stability extended to NA activity. The hVIC/11 mutants containing N93 demonstrated a remarkable 6-fold increase in sialidase activity compared to hVIC/11 in the absence of Ca2+ (hVIC/11-G93N, 22.14%; hVIC/11-A138S/G93N, 31.02%; hVIC/11, 3.8%; Fig. 4f, g). Conversely, the G93 mutation in sOH/04-N93G led to a reduction in activity compared to the sOH/04 control. These contrasting effects suggest that residue 93 not only modulates stability but also plays a crucial role in fine-tuning NA activity in response to Ca2+ availability.
Amino acids 93 and 113 differentially modulate neuraminidase enzymatic parameters
The impact of amino acids 93 and 113 on NA enzymatic activity was investigated by analyzing substrate affinity and sialic acid hydrolysis velocity. Recombinant NA constructs (rNA) consisting of the NA ectodomain fused to various tags (Fig. 5a) were expressed, purified, and evaluated for purity (Supplementary Fig. 8a). Upon cleavage of the introduced domains (Fig. 5b, Supplementary Fig. 8b and c), the released NA ectodomain was used to assess reaction velocity using MUNANA. While D113A did not affect the velocity of hVIC/11-D113A rNA, it reduced the reaction velocity in sOH/04-D113A rNA (Fig. 5c, d). Conversely, introduction of N93 increased the reaction velocity five-fold in hVIC/11-G93N rNA compared to hVIC/11 rNA (0.017 μM/min and 0.0033 μM/min, respectively), while G93 decreased the velocity of sOH/04-N93G rNA compared to sOH/04 rNA (0.011 μM/min versus 0.036 μM/min, Fig. 5d). These findings suggest that residues 93 and 113, although not part of the active site, influence NA catalytic properties.
a Schematic representation of NA constructs expressed in High 5 cells infected with recombinant baculoviruses. b Western blot of recombinant NAs detected using an anti-His antibody before and after thrombin digestion to release the NA ectodomain. c NA enzymatic reaction velocity using recombinant proteins from hVIC/11rNA (yellow), hVIC/11-G93N rNA (brown), hVIC/11-D113A rNA (black), sOH/04 rNA (blue), sOH/04-N93G rNA (orange), and sOH/04-D113A rNA (pink). Samples were normalized to 50 ng of protein and velocity was measured as 4-MU production over 60 min. Values represent the mean ± SD of three independent experiments. d rNA reaction velocity of hVIC/11 rNA (yellow), hVIC/11-G93N rNA (red), hVIC/11-D113A rNA (black), sOH/04 rNA (blue), sOH/04-N93G rNA (orange), and sOH/04-D113A rNA (pink). Values are shown as the means of three independent experiments and p values were obtained by ordinary one-way ANOVA. e Michaelis-Menten plot of rNA samples normalized to 50 ng of protein and incubated with variable MUNANA concentrations. Data was fitted to the Michaelis-Menten equation and the KM (f) and Vmax (g) parameters. were obtained. p values were obtained by ordinary one-way ANOVA. h Thermostability profiles of rNA measured with (2 mM Ca2+, solid line) or without (dashed line) Ca2+. Values are presented as the mean ± SD of three independent experiments. (i) T50 values were obtained by fitting the data to a dose-response variable slope nonlinear fit. p values were obtained by ordinary one-way ANOVA. j rNA enzymatic activity at 0 mM Ca2+, measured at 37 °C. p values were obtained by ordinary one-way ANOVA. k Pearson correlation test between rNA activity and T50 at 0 mM Ca2+.
Further analysis of substrate affinity using the Michaelis constant (KM) revealed that neither G93N nor D113A affected the KM of hVIC/11-G93N rNA or hVIC/11-D113A rNA compared to hVIC/11 rNA (Fig. 5e, f). Conversely, both sOH/04-N93G rNA and sOH/04-D113A rNA exhibited increased KM, indicating reduced substrate affinity compared to sOH/04 rNA. Assessment of maximum velocity (Vmax) showed that hVIC/11-G93N rNA displayed the highest Vmax among hVIC/11-derived NAs, while hVIC/11-D113A rNA had a reduced Vmax compared to hVIC/11 rNA (Fig. 5g). Similarly, G93 and A113 reduced Vmax in sOH/04 NA. These results suggest that residue N93 enhances the reaction Vmax, whereas A113 decreases it.
Calculation of the turnover number (kcat) further supported these observations (Supplementary Fig. 8d, e, Supplementary Table 2), demonstrating increased kcat in hVIC/11-G93N rNA and decreased kcat in hVIC/11-D113A rNA compared to hVIC/11 rNA. The opposite effect was observed in sOH/04 NA mutants. Additionally, the specificity constant (kcat/KM) was increased in hVIC/11-G93N rNA and reduced in both hVIC/11-D113A rNA and sOH/04 NA mutants (Supplementary Fig. 8f, Supplementary Table 2). This analysis indicates that N93 modulates catalytic power by affecting interactions after enzyme-substrate complex formation, while A113 decreases the catalytic efficiency noted as a reduced kcat/KM.
Examination of T50 values linked the increased activity of hVIC/11-G93N rNA to enhanced thermostability, whereas hVIC/11-D113A rNA exhibited reduced T50 under 2 mM Ca2+ (Fig. 5h, i). Similar trends were observed in sOH/04 NA mutants. Notably, both hVIC/11-G93N rNA and hVIC/11-D113A rNA displayed increased stability and activity at 0 mM Ca2+ compared to hVIC/11 rNA (Fig. 5j, Supplementary Fig. 9), whereas sOH/04r-N93G rNA showed reduced activity. The correlation between NA thermostability and activity under Ca2+-depleted conditions was confirmed by a Pearson test (Fig. 5k). Thus, residues 93 and 113 differentially modulate NA catalytic power by influencing the tetramer stability. Notably, amino acids destabilizing Ca2+ binding in the low-affinity Ca2+-binding pocket, prevalent in recent swine H3N2 viruses, enhance NA stability and activity in Ca2+-depleted conditions.
Amino acids 93 and 113 impact the HA/NA balance
To assess the impact of NA mutations at positions 93 and 113 on the balance between HA and NA activities, a red blood cell elution assay was performed, measuring the time required for a 50% reduction in HA titers under NA activity (Fig. 6a–f). At 2 mM Ca2+, hVIC/11-A138S exhibited the fastest elution, while mutations at either residue 93 (G to N) or 113 (D to A) increased elution time. Similarly, sOH/04-N93G showed slower elution kinetics. In the absence of Ca2+, hVIC/11 and hVIC/11-A138S displayed a marked increase in elution time, which was reversed by introducing either N93 or A113. Conversely, sOH/04-N93G maintained a slower elution time even without Ca2+. No elution was observed in the presence of oseltamivir, confirming assay functionality. These differences were statistically significant, indicating that residues 93 and 113 modulate viral elution time, reflecting alterations in the HA/NA balance. The observed changes in elution kinetics suggest that these residues influence the functional interplay between HA and NA, highlighting their role in optimizing viral fitness and adaptation.
Viruses were normalized to 32 HAU and incubated at 4 °C for 1 h before incubation at 37 °C for the indicated time. a Elution kinetics of hVIC/11 mutants at 2 mM Ca2+. b Elution kinetics sOH/04 mutants at 2 mM Ca2+. c Elution kinetics of hVIC/11 and sOH/04 mutants at 2 mM Ca2+ and 1 μM oseltamivir. d Elution kinetics of hVIC/11 mutants at 0 mM Ca2+. e Elution kinetics sOH/04 mutants at 0 mM Ca2+. f Elution kinetics of hVIC/11 and sOH/04 mutants at 0 mM Ca2+ and 1 μM oseltamivir. g Elution time of hVIC/11 and sOH/04 mutants at 2 mM Ca2+. h Elution time of hVIC/11 and sOH/04 mutants at 0 mM Ca2+. Values are presented as the mean ± SD of two independent experiments. p values were determined by one-way ANOVA with Tukey’s multiple comparison test.
Disruption of Ca2+ binding in the low-affinity site reduces Ca2+ dependency for viral replication and aerosol infection
Given the importance of NA activity in the FLUAV replication cycle, the biological significance of the D113A mutation in hVIC/11-A138S/D113A was assessed through plaque assays under varying Ca2+ concentrations (Supplementary Fig. 10). At both 2 mM and 0 mM Ca2+, hVIC/11-D113A and hVIC/11-A138S/D113A produced the largest plaques (Fig. 7a). sOH/04 plaques were larger than those of hVIC/11. The hVIC/11 strain exhibited strong Ca2+ dependency, with plaque size reduced by nearly 60% in the absence of Ca2+ compared to the 2 mM condition (Fig. 7b). Conversely, Ca2+ concentration had less impact on hVIC/11-A138S, hVIC/11-D113A, and sOH/04, with plaque size reductions around ≤40%. The hVIC/11-A138S/D113A mutant showed minimal plaque size reduction under 0 mM Ca2+.
a MDCK cells were infected with 10-fold dilutions of hVIC/11 NA mutants and the control sOH/04 virus in media containing 2 (orange), 0.5 (cyan), or 0 mM Ca2+ (purple). At 72 hpi plaque size diameter was measured for each condition. p values were obtained by ordinary one-way ANOVA. b Effect of Ca2+ concentration on plaque size represented as the percentage of the plaque diameter compared to the plaque size at 2 mM Ca2+ that was arbitrarily set to 100%. p values were obtained by ordinary one-way ANOVA. c Viral particle release assay showing the HA units in 50 μL of supernatant of MDCK cells infected with hVIC/11, sOH/04, hVIC/11-A138S, hVIC/11-D113A, and hVIC/11-A138S/D113A. Cells were infected at an MOI of 0.1 and at 5 hpi the media was replaced with fresh media containing the mentioned Ca2+ concentrations. Values represent the mean ± SD of three independent experiments. d Viral growth kinetics of hVIC/11, sOH/04, hVIC/11-A138S, hVIC/11-D113A, and hVIC/11-A138S/D113A in aerosol infected MDCK cells. Viruses were aerosolized with (orange) or without (purple) Ca2+ and viral titers were recorded at 0, 12, 24, 48, and 72 hpi. Values represent the mean ± SD of three independent experiments. e Viral growth kinetics of hVIC/11, sOH/04, hVIC/11-A138S, hVIC/11-D113A, and hVIC/11-A138S/D113A in aerosol infected PK15 cells. Viruses were aerosolized with (orange) or without (purple) Ca2+ and viral titers were recorded at 0, 12, 24, 48, and 72 hpi. Values represent the mean ± SD of three independent experiments.
Viral particle release, predominantly mediated by NA activity, was also evaluated. The hVIC/11 and hVIC/11-A138S viruses showed decreased release in the absence of Ca2+ (Fig. 7c). In contrast, sOH/04 and hVIC/11-A138S/D113A, particularly the latter, exhibited negligible differences among tested Ca2+ concentrations. These findings suggest that impairing Ca2+ binding in the symmetry axis reduces optimal Ca2+ requirements for growth, likely due to enhanced NA activity under low Ca2+ conditions, leading to improved viral release. This effect was particularly pronounced in aerosol infectivity assessments. The hVIC/11 and hVIC/11-A138S viruses showed delayed kinetics in MDCK cells when aerosolized in Ca2+-free media, reaching lower titers at 72 hpi compared to Ca2+-supplemented media (Fig. 7d). In contrast, hVIC/11-D113A, hVIC/11-A138S/D113A, and sOH/04 showed no differences under both conditions, indicating that viruses with Ca2+-destabilizing mutations are less sensitive to Ca2+ concentrations during aerosolization. This phenomenon was even more evident in swine-origin PK15 cells. While no significant differences were observed among viruses upon direct inoculation (Fig. 3a), aerosol delivery revealed a striking contrast (Fig. 7e). The hVIC/11 strain exhibited minimal replication with Ca2+-supplemented media and no detectable titers without Ca2+. However, hVIC/11-D113A and hVIC/11-A138S/D113A replicated similarly under both conditions, although hVIC/11-D113A titers were ultimately lower. These results demonstrate that viruses with reduced Ca2+ requirements for replication (hVIC/11-A138S/D113A, hVIC/11-D113A, and sOH/04) exhibit reduced dependency on this ion during aerosol infection, with minimal differences in growth kinetics between Ca2+ conditions.
Discussion
The NA protein plays a critical role in FLUAV infection, facilitating viral movement through respiratory mucus and promoting viral release by hydrolyzing sialic acid32,33. NA activity is strongly dependent on Ca2+ 34,35, with some subtypes binding up to two Ca2+ ions per monomer plus a central ion in the symmetry axis36. However, most NA subtypes, including N2, primarily bind two Ca2+ ions in distinct high- and low-affinity binding pockets17. The high-affinity pocket is consistently occupied and essential for NA catalysis, as Ca2+ binding repositions the catalytic residues for proper sialic acid binding26. Recent studies have revealed that the low-affinity pocket’s role in modulating N1 NA activity by stabilizing the tetramer is crucial for optimal function28,37. Findings in this report extend this understanding to N2 FLUAV, demonstrating that the low-affinity pocket also modulates N2 NA catalytic properties, potentially serving as a mechanism for regulating the balance between HA and NA activities, and providing a mechanism to modulate NA enzymatic properties without mutating the catalytic residues.
Leveraging a swine sustained transmission model and a mutant virus with a mutation in HA (hVIC/11-A138S) previously detected after transmission of a human strain to pigs8, we identified a potentially compensatory mutation, D113A, in the N2 NA low-affinity Ca2+-binding pocket. N2 crystal structures of prototypical human strains revealed that residue 113 is the sole amino acid involved in Ca2+ binding at this site in this FLUAV subtype. This contrasts with N1 NAs, where residues K111, D113, and particularly Y170 play key roles in Ca2+ binding in the symmetry axis28. Unlike the 2009 pandemic H1N1 virus, acquisition of D113A by the human-origin N2 NA did not affect viral replication in the cell lines tested. Furthermore, the failure of hVIC/11-D113A to replicate in swine respiratory cells suggests that this mutation alone does not enhance replication in pigs (Fig. 3a), underscoring the critical role of the HA mutation A138S in adaptation to pigs due to increased affinity for swine cells38,39,40. These findings argue for a sequential adaptation model for human FLUAVs in pigs: initial acquisition of adaptive HA mutations to facilitate binding to swine cells, followed by NA mutations, such as D113A, to restore HA/NA balance disrupted by the prior HA changes. This model emphasizes the dynamic interplay between HA and NA in viral adaptation to new hosts, highlighting the importance of compensatory mechanisms in maintaining viral fitness during host transitions.
While the D113A mutation did not affect viral fitness in vitro in this study, it did impact NA thermostability, which is closely linked to Ca2+ binding41. The importance of Ca2+ in enhancing N2 activity has been previously established42. Notably, the D113A mutation increased N2 thermostability under Ca2+-depleted conditions, accompanied by an increase in its activity. Interestingly, swine-adapted N2 NAs inherently exhibit higher sialidase activity in the absence of Ca2+, contrasting with human viruses. The observation of stronger Ca2+ dependency in human-origin N2 aligns with previous reports28,42. The high activity of swine FLUAVs under Ca2+-depleted conditions, despite carrying D113, suggests a lack of Ca2+ binding in the low-affinity Ca2+-binding pocket. Modeling of sOH/04 NA confirmed this hypothesis, revealing that Ca2+ binding is prevented by repositioning of water molecules in the symmetry axis due to a change in D113 side chain location (Fig. 4). Further support for this mechanism was provided by the introduction of D113A in sOH/04 NA, which resulted in no changes in thermostability compared to sOH/04 (Figs. 4 and 5), confirming that the virus does not bind Ca2+ when either amino acid is present. This suggests that the presence of D113, even without direct Ca2+ interaction, contributes to the overall stability of the NA structure, highlighting the complex interplay between amino acid residues and Ca2+ binding in modulating NA function.
Residue 93 appears to be under different selective pressures in humans and swine. While K93 and N93 were present in both human and swine FLUAV in the early 1990s, human isolates rapidly transitioned to D93 and G93, while N93 was retained in swine H3N2 FLUAVs. Protein modeling suggests that the N93 residue destabilizes the interaction of D113 with Ca2+, thereby preventing Ca2+ binding. Interestingly, a similar mechanism has been reported for residue 106 in N1 NAs, which also modulates Ca2+ binding in the low-affinity Ca2+-binding pocket28. This suggests a conserved mechanism between N1 and N2 NAs for modulating Ca2+ binding at this site, involving specific residues distant from the pocket itself. The presence of this mechanism across subtypes may be due to the high conservation of the low-affinity Ca2+-binding pocket. Such a mechanism could prevent modifications near the Ca2+-binding pocket that might interfere with tetramer formation, as NA oligomerization is driven by both the head and transmembrane domains43. However, further investigation is needed to confirm this hypothesis.
Residues 93 and 113 exert similar effects on Ca2+ binding in both human and swine N2 FLUAVs, yet their impact on NA catalytic properties differs. Notably, while D113A minimally affects sOH/04 rNA Vmax, it reduces hVIC/11 rNA velocity. Conversely, N93 drastically increases reaction velocity within the hVIC/11 rNA. Additionally, sOH/04 rNA displays higher substrate affinity than hVIC/11 rNA, with no difference observed in hVIC/11-G93N rNA, suggesting that N93 seems to influence steps occurring after the enzyme-substrate complex formation, while A113 decreases the catalytic efficiency of the enzyme. This aligns with previous reports of Ca2+ modulating reaction velocity by favoring enzyme-substrate interactions44,45, although the mechanism remains unclear. The D113A mutation impacts substrate binding, decreasing affinity regardless of NA backbone, potentially explaining its rarity in natural isolates due to disruption of HA/NA balance46,47. Previous work has demonstrated that hVIC/11-A138S exhibits higher affinity for α2,6-SA, indirectly enhancing NA activity9. Based on this, it was hypothesized that the D113A mutation emerged in vivo to compensate for the A138S change in HA and regulate NA activity. Indeed, data revealed a higher elution rate for hVIC/11-A138S compared to hVIC/11 and sOH/04, which was restored to sOH/04-like levels upon introduction of D113A (Fig. 6). These findings underscore the intricate interplay between HA and NA, whereby mutations in one protein can necessitate compensatory changes in the other to maintain optimal viral fitness. Notably, A113 negatively impacted tetramer stability in the hVIC/11 NA backbone, suggesting that other residues might prevent this effect in sOH/04 NA. These two amino acids thus represent distinct mechanisms for N2 NA modulation of activity during host jumps, explaining the tendency of human viruses to bind Ca2+ in the low-affinity Ca2+-binding pocket, while swine H3N2 viruses do not. This suggests a novel role for Ca2+ in FLUAV host range, where viral changes may modulate sialidase activity without modifying the conserved catalytic pocket, by influencing Ca2+ binding in the low-affinity site. This is supported by previous studies on the 2009 pandemic H1N1 NA crystal structures28. Early isolates, upon jumping from pigs to humans carried V106 that destabilized the tetramer at 2 mM Ca2+, but it was quickly replaced by I106 that enhanced N1 NA thermostability and activity under this condition. While that study did not directly link FLUAV host range and Ca2+ binding, the order of events aligns with the hypothesis that HA changes are required early in host adaptation, followed by NA activity modification through altered substrate affinity or reaction velocity, depending on specific residues involved in Ca2+ binding. However, this process seems to cycle between binding and not binding calcium in the symmetry axis28, which could represent a mechanism to compensate for changes in the HA, as the D113A mutation alone cannot grant replication in swine airway cells (Fig. 3).
Increased NA thermostability conferred by the D113A substitution reduced the Ca2+ requirement for optimal virus replication to levels comparable to sOH/04. This is likely due to enhanced NA activity under low Ca2+ conditions, leading to more efficient virus release from infected cells28. The increased stability of A113-containing mutants also resulted in more efficient aerosol infection of MDCK cells when the inoculum was aerosolized in the absence of Ca2+, similar to sOH/04. This effect was even more pronounced in PK15 cells, with limited to no replication observed for hVIC/11 and hVIC/11-D113A. Notably, hVIC/11-A138S/D113A demonstrated similar replication kinetics to sOH/04 across all tested conditions. These findings suggest that the absence of Ca2+ in the symmetry axis enhances aerosol infection under Ca2+-depleted conditions, potentially reflecting differences in mucus composition between human and swine, which could select for viruses with distinct aerosol stability. This could explain why hVIC/11 fails to transmit in pigs9 and only replicates in the upper respiratory tract of swine (~37 °C) but not in the lungs (~39 °C)8,9, given that the NA of hVIC/11 is almost inactive at 39 °C in the absence of calcium (Figs. 3, 4, and 5). However, further investigation is needed to confirm this hypothesis.
Overall, this study demonstrates distinct Ca2+ binding profiles in human and swine H3N2 FLUAVs, with swine strains lacking Ca2+ binding in the low-affinity pocket. This difference leads to enhanced stability and activity of swine-origin FLUAVs under Ca2+-depleted conditions compared to human-origin FLUAVs, which are almost inactivated under these conditions. Residue 93 modulates Ca2+ binding and significantly impacts reaction velocity by affecting steps after SA binding in the NA catalytic pocket. This difference in reaction velocity and Ca2+ binding suggests a role of Ca2+ in the host range of FLUAV, with those from humans requiring higher Ca2+ concentrations for replication while their swine counterparts have reduced Ca2+ requirements. A better understanding of the biochemical differences of the upper and lower respiratory tract of humans and pigs is needed to understand why swine N2s need to retain high activity under calcium-depleted conditions. Nonetheless, the identification of residues that lead to enhanced fitness in swine cells (such as N93 and A113) can help recognize new spillover events that are more likely to be sustained in pigs and help improve biosecurity measures to decrease viral spread and prevent establishment of novel lineages. These results provide new insights into the mechanisms utilized by FLUAV to regulate NA activity during host adaptation and the potential effects on the HA/NA balance.
Materials and methods
Ethics statement
Animal studies were approved by the Institutional Care and Use Committee (IACUC) at the University of Georgia (protocol A2019 03-031-Y3-A9). Studies were performed under animal biosafety level 2+ (ABSL2+) containment and animals were cared for in accordance with the guidelines set forth in the “Guide for the Care and Use of Agricultural Animals in Research and Teaching (Ag Guide)” (American Dairy Science Association, American Society of Animal Science, and Poultry Science Association, 2020). At the end of the study, animals were euthanized following the American Veterinary Medical Association (AVMA) guidelines. All experiments with mutant viruses were performed under BSL 2+ conditions at University of Georgia (Institutional Biosafety Committee approval number 2020-0035).
Cells and viruses
Madin-Darby canine kidney (MDCK), human lung carcinoma (A549), and porcine kidney (PK15) cells were grown in Dulbecco’s Modified Eagles Medium (DMEM, Sigma-Aldrich, St Louis, MO) supplemented with 2 mM L-glutamine (Sigma-Aldrich, St Louis, MO), 10% fetal bovine serum (FBS, Sigma-Aldrich, St Louis, MO), and 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO). Cells were cultured at 37 °C in a humidified incubator with 5% CO2. MDCK cells were a kind gift from Dr. Robert Webster (St Jude Children’s Research Hospital, SJCRH). A549 and PK15 cells were purchased from the American Type Culture Collection (ATCC, Manassas, VA).
Human airway epithelial cells (BCi.NS1.1)48 were kindly provided by Dr. Ronald Crystal (Weill Cornell Medicine, New York) and maintained in PneumaCult-Ex Plus Basal Media (STEMCELL Technologies, Vancouver, Canada) supplemented with PneumaCult-Ex Plus Supplement (STEMCELL Technologies, Vancouver, Canada), 0.1% hydrocortisone (STEMCELL Technologies, Vancouver, Canada), 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO), and 0.5% gentamycin (Sigma-Aldrich, St Louis, MO). Cells were cultured at 37 °C in a humidified incubator with 5% CO2 and media was replaced every two days. Differentiation of BCi.NS1.1 cells was performed in type IV collagen-coated 12 mm trans well plates (0.4 μm pore size, Corning Inc., NY, USA). Cells were plated at 3 × 105 cells/well and cultures were maintained at 37 °C in a humidified incubator with 5% CO2 until a 100% confluency was reached. After reaching confluency, cells were differentiated by changing to air-liquid interface (ALI) conditions by removing the apical media and replacing the basal media with PneumaCult ALI Base Media (STEMCELL Technologies, Vancouver, Canada), supplemented with PneumaCult ALI Supplement (STEMCELL Technologies, Vancouver, Canada), 1% PneumaCult ALI Maintenance Supplement (STEMCELL Technologies, Vancouver, Canada), 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO), and 0.5% gentamycin (Sigma-Aldrich, St Louis, MO), 0.2% heparin (STEMCELL Technologies, Vancouver, Canada), and 0.5% hydrocortisone (STEMCELL Technologies, Vancouver, Canada). Cells were maintained at 37 °C in a humidified incubator with 8% CO2 for 5 days and then were cultured at 37 °C in a humidified incubator with 5% CO2 for 16 more days.
Spodoptera frugiperda pupal ovarian tissue (Sf9) cells were purchased from ThermoFisher Scientific (Waltham, MA) and maintained in SF-900 II SFM media (ThermoFisher Scientific, Waltham, MA) supplemented with 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO). High Five™ Insect cells (BTI-TN-5B1-4) were obtained from ThermoFisher Scientific (Waltham, MA) and cultured in Express Five SFM media (ThermoFisher Scientific, Waltham, MA) supplemented with 18 mM L-glutamine (ThermoFisher Scientific, Waltham, MA) and 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO). Both insect cell lines were maintained in suspension at 28 °C.
FLUAV viruses used in this study (Table 1) were generated using an eight-plasmid reverse genetics system as previously described49. Parental viruses (hVIC/11, sOH/04, and hVIC/11-A138S) were described before8,9. D113A and G93N/N93G mutations were introduced into hVIC/11 or sOH/04 NA by site-directed mutagenesis using the Phusion site-directed mutagenesis kit (ThermoFisher Scientific, Waltham, MA), and the primers listed in Supplementary Table 3 according to the manufacturer’s instructions and plasmids’ sequences were confirmed by whole plasmid sequencing. Viruses were propagated in MDCK cells using Opti-MEM (ThermoFisher Scientific, Waltham, MA) containing 1 μg/ml of tosylsulfonyl phenylalanyl chloromethyl ketone (TPCK)-treated trypsin (Worthington Biochemicals, Lakewood, NJ) at 37 °C. Viral titers were determined by TCID50 using the Reed and Muench method50.
In vivo studies
3-weeks-old cross-bred pigs obtained from Midwest Research Swine Inc (Glencoe, MN, USA) and housed in animal biosafety level 2 (ABSL2) facilities at the University of Georgia. Animals were negative for anti-FLUAV NP antibodies by competitive ELISA (IDEXX, Westbrook, ME) and randomly distributed into two groups. Pigs (n = 3, seeders) were inoculated intratracheally and intranasally under anesthesia using a cocktail of ketamine (6 mg/kg), xylazine (3 mg/kg), and Telazol (6 mg/kg) with 3 × 106 TCID50 per pig of hVIC/11-A138S or sOH/04. Animals were checked daily for clinical signs and nasal swabs were collected at 0, 1, 2, 3, and 5 days-post infection (dpi) in 2 mL of brain hart infusion broth (Sigma-Aldrich, St Louis, MO). At 2 dpi, naïve pigs (n = 3) were placed as contacts (contact 1, C1) with the directly inoculated animals and were kept together until 5 dpi. At 5 dpi, seeders pigs were sedated using a cocktail of ketamine (3 mg/kg), xylazine (1.5 mg/kg), and Telazol (3 mg/kg) and then humanely euthanized by an intravenous pentobarbital overdose (Euthasol, 390 mg/10 lb). Contact 1 pigs were moved into a clean enclosure after removal of seeder pigs, and additional naïve pigs (n = 3) were placed as new contacts (contact 2, C2). This process was repeated for a total of 4 contacts. Nasal swabs from contact pigs were collected at 0, 1, 3, 5, and 6 days-post contact (dpc). At 6 dpc, contact pigs were sedated and humanely euthanized as described above. During necropsies, bronchoalveolar lavage fluid (BALF) and lung tissue were collected from all pigs.
Virus titration from lung tissue, nasal swabs, and BALF samples
Lung samples were homogenized using the Tissue Lizer II (Qiagen, Gaithersburg, MD) and a Tungsten carbide 3 mm bead (Qiagen, Gaithersburg, MD) for 10 min at 30 Hz. Subsequently, RNA from lung tissues, nasal swabs, and BALF samples was extracted using the MagMax-96 AI/ND viral RNA (ThermoFisher Scientific, Waltham, MA) isolation kit according to manufacturer’s instructions. RNA from lung tissue was normalized to 1 μg in 20 μL of nuclease-free water while RNA from nasal swabs and BALF samples were used directly for titration. Samples were titrated using the Quantabio qScript XLT One-Step RT-qPCR ToughMix kit (Quantabio, Beverly MA) targeting the M segment (Supplementary Table 3) according to manufacturer’s instructions.
Primary swine tracheal epithelial cells isolation
Trachea samples were obtained from three 7-weeks old cross-bred pigs obtained from Midwest Research Swine Inc (Glencoe, MN) and airway epithelial cells were isolated as previously described51 with minor modifications. Briefly, trachea sections were digested for 12 h at 4 °C in DMEM/F12 (Sigma-Aldrich, St Louis, MO) supplemented with 1.5 mg/mL pronase (Sigma-Aldrich, St Louis, MO), and 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO). After overnight incubation, trachea sections were scraped to release individual cells, and fibroblasts were discarded by plastic adherence for 4 h52. Non-adherent cells were then plated in type I collagen-coated flasks and cultured with BEGMTM Bronchial Epithelial Cell Growth Medium BulletKitTM (Lonza, Bend, OR, USA) at 37 °C in a humidified incubator with 5% CO2. Differentiation was performed by seeding 3 × 105 cells/well in type I collagen-coated 12 mm transwell plates (0.4 μm pore size, Corning Inc., NY, USA) in BEGMTM media at 37 °C in a humidified incubator with 5% CO2 until a 100% confluency was reached. After reaching confluency, BEGMTM media was removed from the apical and basal compartments and ALI media composed of DMEM/F12 supplemented with 2% NuSerum (Corning Inc., NY, USA), 2mM L-glutamine (Sigma-Aldrich, St Louis, MO), 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO), and 15 ng/mL retinoic acid (Sigma-Aldrich, St Louis, MO) was added to the basal compartment. ALI media was changed every two days and ALI conditions were maintained for 3 weeks prior to use.
Next-generation sequencing
Viral genomic RNA was extracted from nasal swabs collected at 1, 2, and 5 dpi or 1, 3, and 6 dpc and BALF samples using the MagMax-96 AI/ND viral RNA isolation kit (ThermoFisher Scientific, Waltham, MA) according to manufacturer’s instructions. After extraction, HA and NA segments were amplified using specific primers (Supplementary Table 3) and the SuperScript III One-Step PCR System (ThermoFisher Scientific, Waltham, MA) according to manufacturer’s instructions in duplicates. Duplicate PCR products from both HA and NA PCRs from the same sample were pooled in equal volumes and cleaned with 0.45X Agencourt AMPure XP Magnetic Beads (Beckman Colter, Brea, CA, USA) according to manufacturer’s instructions. Clean PCR product concentration was measured using the Qubit dsDNA HS assay kit (ThermoFisher Scientific, Waltham, MA) on a Qubit 3.0 fluorometer (ThermoFisher Scientific, Waltham, MA) and normalized to 0.2 ng/μL. Sequencing libraries were prepared with the Nextera XT DNA library preparation kit (Illumina, San Diego, CA, USA). Indexed libraries were cleaned with 0.7X Agencourt AMPure XP Magnetic Beads and samples size distribution was analyzed using the Agilent Bioanalyzer 2100 DNA-HS assay (Agilent, Santa Clara, CA, USA). Libraries were then pooled, denatured, and further diluted to 10 pM. Finally, samples were sequenced using a MiSeq Reagent Kit V2, 300 cycles (Illumina, San Diego, CA, USA).
Variant analysis
Analyses of FLUAV variants were performed as previously described53,54. Briefly, sequencing adapters were removed using Cutadapt (version 3.4) and then mapped back to their respective reference sequence using option mem from BWA (version 0.7.17)55. Read-pairing errors that may have been introduced by the alignment were corrected using option fixmate, and secondary and unmapped reads were removed. The resulting bam file was used by LoFreq56 following practices outlined in the Genome Analysis Toolkit57 to identify non-consensus variants. Only variants with a frequency of 0.01 (1%) and coverage equal to or above 100 were used. Synonymous and nonsynonymous mutations were identified using SNPdat (version v1.0.5)58. The output from SNPdat was inputted into RStudio (R version 4.1.2) for visualization. Variants with a frequency equal to or above 0.5 (50%) were considered major variants.
Diversity analysis
Nucleotide diversity π was calculated using the πN (nonsynonymous) to πS (synonymous) ratio statistics determined using SNPGenie59 for estimation of nucleotide diversity from next-generation sequencing. An arbitrary minimal variant frequency of 0.01 was set for π determination.
Amino acid temporal frequency analysis
55,590 human origin and 15,514 swine origin N2 sequences from H3N2 FLUAVS from 1992 to 2022 were obtained from GISAID60. Duplicated sequences were discarded, and samples were aligned using Clustal Omega version 1.2.261. Alignments were exported and amino acid frequency at position 93 and 113 was assessed using Geneious version 2024.0.2.
NA homology model
FLUAV NA sialidase domains were modeled using the SWISS-MODEL server (https://swissmodel.expasy.org/)62. hVIC/11 NA model was made using A/Tanzania/205/2010 as template (PDB accession code: 4GZO63) while A/Moscow/10/1999 (PDB accession code: 8DWB64) was used to generate the sOH/04 NA 3D structure. NA structures were visually inspected using PyMOL 3.0 and model quality was assessed using PROCHECK65.
Growth kinetics
MDCK, A549, and PK15 cells were seeded in 6 well-plates and incubated at 37 °C in a humidified incubator with 5% CO2 until an 80% confluency before use. Cells were infected at a MOI of 0.01 for 1 h at 37 °C. After incubation, cells were washed three times with PBS and supplemented with fresh Opti-MEM containing 1 μg/ml TPCK-treated trypsin. Timepoints were collected at 0, 12, 24, 48, and 72 hpi, and viral titers were determined by RT-qPCR. HAE and differentiated swine tracheal cells were infected by adding 200 μL of virus in the respective ALI media into the apical compartment and incubated for 1 h at 37 °C. After incubation, cells were washed 5 times with PBS. Timepoints were collected at 0, 12, 24, 48, and 72 hpi by adding 200 μL of PBS onto the cells and incubating them at 37 °C for 15 min. Viral titers were determined as described above.
Neuraminidase activity and thermostability
Neuraminidase sialidase activity was evaluated using the whole virus as previously described66 or purified recombinant NA (rNA). Briefly, viruses were diluted in reaction buffer (32.5 mM 2-(N-morpholino)ethanesulfonic acid -MES-, 2 mM CaCl2 -unless stated otherwise-, pH 6.5) and incubated at 37 °C for 30 min. To evaluate purified recombinant proteins, samples were normalized to 50 ng for all of the assays described below. After incubation, 100 μM 2′-(4-Methylumbelliferyl)-α-D-N-acetylneuraminic acid sodium salt hydrate (MUNANA, Sigma-Aldrich, St Louis, MO) was added and reactions were incubated for 60 min at 37 °C. Fluorescence was measured every 60 s at excitation and emission wavelengths of 360 nm and 460 nm, respectively, using a Synergy HTX Multi-Mode Microplate Reader (Agilent BioTek, Santa Clara, CA).
NA thermostability was assessed by incubating equal amounts of each virus at 33, 37, 40, 43, 46, 49, 52, 55, 58, 61, and 64 °C for 10 min. After incubation, viruses’ NA sialidase activity was measured as mentioned above. NA relative activity was calculated as percentage of the activity at 33 °C. T50 values were obtained by adjusting the curves to a non-linear dose-response logarithmic four parameters equation of the form:
Where Y is NA activity in percentage, X is the log(temperature), a is the theoretical maximum activity, b is the theoretical minimum activity, and H is the Hill slope.
NA sialidase activity under different concentrations of Ca2+ was performed by adding 2, 1.5, 1, 0.5, or 0 mM CaCl2 to the reaction buffer (32.5 mM MES, pH 6.5). Reaction velocity was measured as described above, and relative NA activity was calculated by comparing the reaction velocity under each condition to the 37 °C and 2 mM Ca2+ condition.
Enzyme kinetics
rNA samples were diluted in NA reaction buffer and incubated at 37 °C for 30 min. Then 4-MU production over time was evaluated under different MUNANA concentrations (1.17, 2.34, 4.68, 9.37, 18.75, 37.5, 75, 150, 300, and 600 μM). Fluorescence was measured every 60 s at excitation and emission wavelengths of 360 nm and 460 nm and data was fitted to a simple saturation-kinetics Eq. (1) and kcat was obtained by rearranging the Michaelis–Menten Eq. (2) assuming saturated substrate concentrations and a steady-state model:
Red blood cells elusion assay
Viruses were normalized at 32 HA units (HAU) in Ca2+ -free PBS or PBS supplemented with 2 mM Ca2+. Normalized samples were then mixed with 0.5% turkey red blood cells and incubated at 4 °C for 1 h. Then, samples were incubated at 37 °C, and aliquots were collected every 30 min until all HAU were lost. As a control, a duplicate sample supplemented with 1 μM oseltamivir (Sigma-Aldrich, St Louis, MO) was included. Elution due to NA activity was confirmed by incubation at 4 °C for 1 h to allow re-agglutination.
NA protein expression and purification
NA ectodomain from hVIC/11 and sOH/04 was cloned into the pFastBac1 transfer vector. The stalk domain was replaced and fused in frame to a GG linker, a thrombin cleavage site, a tetramerization domain from the human vasodilator-stimulated phosphoprotein (VASP), 6x His tag, and a secretion signal as previously described67. D113A and G93N/N93G mutations were introduced as described above. Cloned pFastBac1 plasmids were then used to generate the recombinant bacmids by transforming DH10Bac competent cells (ThermoFisher Scientific, Waltham, MA), and insert transposition was confirmed by colony PCR using M13 primers (Supplementary Table 3). Bacmids were purified by alkaline lysis and used to rescue recombinant baculoviruses in Sf9 cells according to the Bac-to-Bac system manual (ThermoFisher Scientific, Waltham, MA).
NA proteins were produced by infecting High Five cells at an MOI of 10. When cells reached an 80% mortality (∼72 hpi), the supernatant was cleared by centrifugation at 3000 rpm. NA constructs were purified from clarified supernatant by immobilized metal affinity chromatography (IMAC) using the HisPur Ni-NTA Spin purification kit (ThermoFisher Scientific, Waltham, MA) according to the manufacturer’s instructions and eluted in elution buffer containing 137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4, 1.8 mM KH2PO4, and 300 mM imidazole. Subsequently, eluted NAs were further purified by size exclusion chromatography using a Superdex-200 (Sigma-Aldrich, St Louis, MO) column with a running buffer composed of 20 mM Tris-HCl, 50 mM NaCl, pH 8.00. NA activity was monitored for each fraction. Finally, purified samples were digested using the Thrombin CleanCleave Kit (Sigma-Aldrich, St Louis, MO) according to the manufacturer’s instructions, and digestion products were passed through a Superdex-200 column.
SDS-PAGE and western blot
Protein concentration was determined using the Pierce BCA Protein Assay kit (ThermoFisher Scientific, Waltham, MA) according to the manufacturer’s instructions. 100 ng of either cleaved or non-cleaved NA constructs were boiled for 5 min and resolved by SDS-PAGE using a Mini-Protean TGX Stain-Free Precast Gel (Bio-Rad, Hercules, CA). For total protein imagining, gels were then fixed with 40% ethanol and 10% acetic acid in water for 1 h. Then samples were sensitized for 1 min with 0.02% Na2S2O3 and incubated for 20 min with staining solution (0.1% AgNO3 and 0.02% formaldehyde in water). Finally, gels were developed by incubating them in 3% NaCO3 and 0.05% formaldehyde in water. Gels used for western blot were transferred to a TransBlot Turbo Mini Nitrocellulose membrane and blocked for 2 h in blocking buffer (5% BSA in PBS) at room temperature. After incubation, membranes were washed two times with T-PBS (0.05% Tween-20 in PBS, washing buffer) and incubated for 1 h with an anti-His tag primary antibody (Sigma-Aldrich, St Louis, MO). Membranes were then washed three times (5 min each) and incubated for 1 h with a secondary HRP-conjugated anti-mouse antibody in a 1:1,000 dilution in T-PBS. Finally, membranes were washed 5 times and then developed with the SuperSignal West Femto Maximun Sensibility Substrate kit (ThermoFisher Scientific, Waltham, MA). Membranes were imaged using a ChemiDoc MP Imaging System (Bio-Rad, Hercules, CA).
Plaque assay
MDCK cells were seeded at a density of 105 cells/cm2 and incubated at 37 °C in a humidified incubator with 5% CO2 in Opti-MEM (Life Technologies, Carlsbad, CA, USA) until a 100% confluency before use. The day of infection, culture media was discarded, cells were washed three times with Ca2+ -free DMEM (ThermoFisher Scientific, Waltham, MA), and were infected using 10-fold serial dilution of each virus in Ca2+ -free DMEM. After 1 h incubation at 37 °C, unbound virus was removed by washing cells three times with Ca2+ -free DMEM and then cells were supplemented with DMEM containing 2 mM L-glutamine, 0.3% bovine serum albumin (BSA, ThermoFisher Scientific, Waltham, MA), 1% antibiotic/antimycotic, 1 mM sodium pyruvate (ThermoFisher Scientific, Waltham, MA), 0.8% avicel, and variable concentrations of Ca2+ chloride (2, 0.5 or 0 mM, Sigma-Aldrich, St Louis, MO). Infections were incubated for 72 h at 37 °C. Finally, cells were fixed with 37% formaldehyde (Sigma-Aldrich, St Louis, MO) and stained for 15 min with 0.5% crystal violet in 20% methanol.
Viral particle release
Viral particles release was evaluated as previously described28. Briefly, MDCK cells were infected at a multiplicity of infection (MOI) of 0.1 and were incubated at 4 °C for 30 min. Unbound virus was removed by washing cells three times with PBS and cells were supplemented with fresh Opti-MEM containing 1 μg/ml TPCK-treated trypsin (Worthington Biochemicals, Lakewood, NJ). At 5 hpi, media was discarded and 3 mL of DMEM containing 2 mM L-glutamine, 0.3% BSA, 1% antibiotic/antimycotic, 1 mM sodium pyruvate, 1 μg/ml TPCK-treated trypsin, and variable concentrations of CaCl2 was added. Starting at 6 hpi, 100 μL was collected from each condition every 1 h until 12 hpi, and HA titers were measured using 50 μL of sample and 50 μL 0.5% turkey red blood cells.
Aerosol infection
MDCK and PK15 cells were seeded and incubated as described above until 80% confluence was reached. Viruses were diluted in Ca2+ -free DMEM supplemented with 2 mM L-glutamine (Sigma-Aldrich, St Louis, MO), 0.3% BSA (Sigma-Aldrich, St Louis, MO), and 1% antibiotic/antimycotic (Sigma-Aldrich, St Louis, MO) and used at an MOI of 0.01. For infections performed with Ca2+, 2 mM CaCl2 was added to the infection media. A total of 6 mL of inoculum was aerosolized using the Aeroneb Lab Neubilizer (Small VMD; Kent Scientific, CT, USA) with an expected particle size of 2.5–4 μm. Aerosol was passed through an exposure chamber at a flow rate of 0.1 mL/min. Cells were exposed for 15 min at room temperature, followed by a 5 min purge. Infections were incubated for 1 h at 37 °C and then washed three times with PBS. Finally, Opti-MEM media supplemented with 1 μg/ml TPCK-treated trypsin was added, and time points were collected at 0, 12, 24, 48, and 72 hpi.
Statistics and reproducibility
All statistical analyses were performed using GraphPad Prism Version 10.3.1. P values were obtained by ordinary one-way ANOVA with Tukey’s multiple comparison test. No data was excluded from the analyses. All data is presented as the mean ± standard deviation. Animal sample size of three animals per group (seeders or contacts) was based on previously published work68,69. Animals were randomly allocated to each group to ensure reproducibility. In vitro experiments were performed at least three times independently in triplicates (n = 3 biological replicates) unless stated otherwise in the figure legend, and results were reproducible between experiments.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
All data relevant for this manuscript is provided as a supplementary data file. Sequencing data is available through NCBI’s short read archive BioProject accession number PRJNA1117070.
Code availability
Custom computer code used in this study are available on GitHub (https://github.com/genferreri/Intra--and-inter-host-evolution-of-H9N2-influenza-A-virus-in-Japanese-quail).
References
Skehel, J. J. & Wiley, D. C. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu Rev. Biochem. 69, 531–569 (2000).
Pabis, A., Rawle, R. J. & Kasson, P. M. Influenza hemagglutinin drives viral entry via two sequential intramembrane mechanisms. Proc. Natl Acad. Sci. USA 117, 7200–7207 (2020).
Cohen, M. et al. Influenza A penetrates host mucus by cleaving sialic acids with neuraminidase. Virol. J. 10, 321 (2013).
Ohuchi, M., Asaoka, N., Sakai, T. & Ohuchi, R. Roles of neuraminidase in the initial stage of influenza virus infection. Microbes Infect. 8, 1287–1293 (2006).
Mair, C. M., Ludwig, K., Herrmann, A. & Sieben, C. Receptor binding and pH stability—how influenza A virus hemagglutinin affects host-specific virus infection. Biochim. Biophys. Acta 1838, 1153–1168 (2014).
Shi, Y., Wu, Y., Zhang, W., Qi, J. & Gao, G. F. Enabling the ‘host jump’: structural determinants of receptor-binding specificity in influenza A viruses. Nat. Rev. Microbiol. 12, 822–831 (2014).
Shelton, H., Roberts, K. L., Molesti, E., Temperton, N. & Barclay, W. S. Mutations in haemagglutinin that affect receptor binding and pH stability increase replication of a PR8 influenza virus with H5 HA in the upper respiratory tract of ferrets and may contribute to transmissibility. J. Gen. Virol. 94, 1220–1229 (2013).
Mo, J. S. et al. Transmission of human influenza A virus in pigs selects for adaptive mutations on the HA gene. J. Virol. 96, e0148022 (2022).
Cardenas, M. et al. Amino acid 138 in the HA of a H3N2 subtype influenza A virus increases affinity for the lower respiratory tract and alveolar macrophages in pigs. PLoS Pathog. 20, e1012026 (2024).
Rajao, D. S. et al. Changes in the hemagglutinin and internal gene segments were needed for human seasonal H3 influenza A virus to efficiently infect and replicate in swine. Pathogens 11. https://doi.org/10.3390/pathogens11090967 (2022).
Rajao, D. S., Vincent, A. L. & Perez, D. R. Adaptation of human influenza viruses to swine. Front. Vet. Sci. 5, 347 (2018).
Rajao, D. S. et al. Novel reassortant human-like H3N2 and H3N1 influenza A viruses detected in pigs are virulent and antigenically distinct from swine viruses endemic to the United States. J. Virol. 89, 11213–11222 (2015).
Nelson, M. I. et al. Introductions and evolution of human-origin seasonal influenza A viruses in multinational Swine populations. J. Virol. 88, 10110–10119 (2014).
Nelson, M. I., Stratton, J., Killian, M. L., Janas-Martindale, A. & Vincent, A. L. Continual reintroduction of human pandemic H1N1 influenza A viruses into Swine in the United States, 2009 to 2014. J. Virol. 89, 6218–6226 (2015).
de Vries, E., Du, W., Guo, H. & de Haan, C. A. M. Influenza A virus hemagglutinin-neuraminidase-receptor balance: preserving virus motility. Trends Microbiol. 28, 57–67 (2020).
Mitnaul, L. J. et al. Balanced hemagglutinin and neuraminidase activities are critical for efficient replication of influenza A virus. J. Virol. 74, 6015–6020 (2000).
McAuley, J. L., Gilbertson, B. P., Trifkovic, S., Brown, L. E. & McKimm-Breschkin, J. L. Influenza virus neuraminidase structure and functions. Front Microbiol 10, 39 (2019).
Guo, Z. et al. Neuraminidase activity modulates cellular coinfection during influenza A virus multicycle growth. mBio 14, e0359122 (2023).
Yang, X. et al. A beneficiary role for neuraminidase in influenza virus penetration through the respiratory mucus. PLoS One 9, e110026 (2014).
Colman, P. M., Varghese, J. N. & Laver, W. G. Structure of the catalytic and antigenic sites in influenza virus neuraminidase. Nature 303, 41–44 (1983).
Bucher, D. J. & Kilbourne, E. D. A 2 (N2) neuraminidase of the X-7 influenza virus recombinant: determination of molecular size and subunit composition of the active unit. J. Virol. 10, 60–66 (1972).
Paterson, R. G. & Lamb, R. A. Conversion of a class II integral membrane protein into a soluble and efficiently secreted protein: multiple intracellular and extracellular oligomeric and conformational forms. J. Cell Biol. 110, 999–1011 (1990).
Li, Q. et al. Structural and functional characterization of neuraminidase-like molecule N10 derived from bat influenza A virus. Proc. Natl Acad. Sci. USA 109, 18897–18902 (2012).
Gong, J., Xu, W. & Zhang, J. Structure and functions of influenza virus neuraminidase. Curr. Med Chem. 14, 113–122 (2007).
Colman, P. M., Hoyne, P. A. & Lawrence, M. C. Sequence and structure alignment of paramyxovirus hemagglutinin-neuraminidase with influenza virus neuraminidase. J. Virol. 67, 2972–2980 (1993).
Smith, B. J. et al. Structure of a calcium-deficient form of influenza virus neuraminidase: implications for substrate binding. Acta Crystallogr. D. Biol. Crystallogr. 62, 947–952 (2006).
Lawrenz, M. et al. Impact of calcium on N1 influenza neuraminidase dynamics and binding free energy. Proteins 78, 2523–2532 (2010).
Wang, H., Dou, D., Ostbye, H., Revol, R. & Daniels, R. Structural restrictions for influenza neuraminidase activity promote adaptation and diversification. Nat. Microbiol. 4, 2565–2577 (2019).
van der Vries, E. et al. H1N1 2009 pandemic influenza virus: resistance of the I223R neuraminidase mutant explained by kinetic and structural analysis. PLoS Pathog. 8, e1002914 (2012).
Li, Q. et al. The 2009 pandemic H1N1 neuraminidase N1 lacks the 150-cavity in its active site. Nat. Struct. Mol. Biol. 17, 1266–1268 (2010).
Pena, L. et al. Modifications in the polymerase genes of a swine-like triple-reassortant influenza virus to generate live attenuated vaccines against 2009 pandemic H1N1 viruses. J. Virol. 85, 456–469 (2011).
Iseli, A. N. et al. The neuraminidase activity of influenza A virus determines the strain-specific sensitivity to neutralization by respiratory mucus. J. Virol. 97, e0127123 (2023).
Sakai, T., Nishimura, S. I., Naito, T. & Saito, M. Influenza A virus hemagglutinin and neuraminidase act as novel motile machinery. Sci. Rep. 7, 45043 (2017).
Baker, N. J. & Gandhi, S. S. Effect of Ca++ on the stability of influenza virus neuraminidase. Arch. Virol. 52, 7–18 (1976).
Dimmock, N. J. Dependence of the activity of an influenza virus neuraminidase upon Ca2+. J. Gen. Virol. 13, 481–483 (1971).
Xu, X., Zhu, X., Dwek, R. A., Stevens, J. & Wilson, I. A. Structural characterization of the 1918 influenza virus H1N1 neuraminidase. J. Virol. 82, 10493–10501 (2008).
Burmeister, W. P., Ruigrok, R. W. & Cusack, S. The 2.2 A resolution crystal structure of influenza B neuraminidase and its complex with sialic acid. EMBO J. 11, 49–56 (1992).
Xu, R. et al. Functional balance of the hemagglutinin and neuraminidase activities accompanies the emergence of the 2009 H1N1 influenza pandemic. J. Virol. 86, 9221–9232 (2012).
Youk, S. S. et al. Mutations in PB1, NP, HA, and NA contribute to increased virus fitness of H5N2 highly pathogenic avian influenza virus clade 2.3.4.4 in chickens. J. Virol. 95, e01675-20 (2021).
Scheibner, D. et al. Phenotypic effects of mutations observed in the neuraminidase of human origin H5N1 influenza A viruses. PLoS Pathog. 19, e1011135 (2023).
Burmeister, W. P., Cusack, S. & Ruigrok, R. W. Calcium is needed for the thermostability of influenza B virus neuraminidase. J. Gen. Virol. 75, 381–388 (1994).
Klenow, L. et al. Influenza virus and pneumococcal neuraminidases enhance catalysis by similar yet distinct sialic acid-binding strategies. J. Biol. Chem. 299, 102891 (2023).
da Silva, D. V., Nordholm, J., Madjo, U., Pfeiffer, A. & Daniels, R. Assembly of subtype 1 influenza neuraminidase is driven by both the transmembrane and head domains. J. Biol. Chem. 288, 644–653 (2013).
Pegg, M. S. & von Itzstein, M. Slow-binding inhibition of sialidase from influenza virus. Biochem. Mol. Biol. Int. 32, 851–858 (1994).
Buxton, R. C. et al. Development of a sensitive chemiluminescent neuraminidase assay for the determination of influenza virus susceptibility to zanamivir. Anal. Biochem. 280, 291–300 (2000).
Shtyrya, Y. et al. Adjustment of receptor-binding and neuraminidase substrate specificities in avian-human reassortant influenza viruses. Glycoconj. J. 26, 99–109 (2009).
Kaverin, N. V. et al. Intergenic HA-NA interactions in influenza A virus: post reassortment substitutions of charged amino acid in the hemagglutinin of different subtypes. Virus Res 66, 123–129 (2000).
Walters, M. S. et al. Generation of a human airway epithelium derived basal cell line with multipotent differentiation capacity. Respir. Res. 14, 135 (2013).
Hoffmann, E., Neumann, G., Kawaoka, Y., Hobom, G. & Webster, R. G. A DNA transfection system for generation of influenza A virus from eight plasmids. Proc. Natl Acad. Sci. USA 97, 6108–6113 (2000).
Reed, L. J. & Muench, H. A simple method of estimating fifty percent endpoints12. Am. J. Epidemiol. 27, 493–497 (1938).
Meliopoulos, V. et al. Primary swine respiratory epithelial cell lines for the efficient isolation and propagation of influenza A viruses. J. Virol. 94. https://doi.org/10.1128/JVI.01091-20 (2020).
Wang, H. et al. Establishment and comparison of air-liquid interface culture systems for primary and immortalized swine tracheal epithelial cells. BMC Cell Biol. 19, 10 (2018).
Siegers, J. Y. et al. Evolution of highly pathogenic H5N1 influenza A virus in the central nervous system of ferrets. PLoS Pathog. 19, e1011214 (2023).
Ferreri, L. M. et al. Intra- and inter-host evolution of H9N2 influenza A virus in Japanese quail. Virus Evolut. 8, veac001 (2022).
Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).
Wilm, A. et al. LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets. Nucleic Acids Res. 40, 11189–11201 (2012).
Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinform. 43, 11 10 11-11 10 33. https://doi.org/10.1002/0471250953.bi1110s43 (2013).
Doran, A. G. & Creevey, C. J. Snpdat: easy and rapid annotation of results from de novo snp discovery projects for model and non-model organisms. BMC Bioinform. 14, 45 (2013).
Nelson, C. W., Moncla, L. H. & Hughes, A. L. SNPGenie: estimating evolutionary parameters to detect natural selection using pooled next-generation sequencing data. Bioinformatics 31, 3709–3711 (2015).
Khare, S. et al. GISAID’s role in pandemic response. China CDC Wkly 3, 1049–1051 (2021).
Sievers, F. et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).
Waterhouse, A. et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 46, W296–W303 (2018).
Zhu, X. et al. Influenza virus neuraminidases with reduced enzymatic activity that avidly bind sialic Acid receptors. J. Virol. 86, 13371–13383 (2012).
Lei, R. et al. Mutational fitness landscape of human influenza H3N2 neuraminidase. Cell Rep. 42, 113356 (2023).
Laskowski, R. A., Rullmannn, J. A., MacArthur, M. W., Kaptein, R. & Thornton, J. M. AQUA and PROCHECK-NMR: programs for checking the quality of protein structures solved by NMR. J. Biomol. NMR 8, 477–486 (1996).
Marathe, B. M., Leveque, V., Klumpp, K., Webster, R. G. & Govorkova, E. A. Determination of neuraminidase kinetic constants using whole influenza virus preparations and correction for spectroscopic interference by a fluorogenic substrate. PLoS One 8, e71401 (2013).
Ellis, D. et al. Structure-based design of stabilized recombinant influenza neuraminidase tetramers. Nat. Commun. 13, 1825 (2022).
Murcia, P. R. et al. Evolution of equine influenza virus in vaccinated horses. J. Virol. 87, 4768–4771 (2013).
Murcia, P. R. et al. Evolution of an Eurasian avian-like influenza virus in naive and vaccinated pigs. PLoS Pathog. 8, e1002730 (2012).
Acknowledgements
The authors thank the University Research Animal Resources personnel at the University of Georgia for assistance with animal care. We thank Dr. Lucas Ferreri for the development of the pipeline used for variant analysis. This research was supported by Agriculture and Food Research Initiative grant no. 2020-67015-31563/project accession no. 1022827 from the USDA National Institute of Food and Agriculture to DSR. Funding was also provided, in part, by The National Pork Board to DSR under Project #21-085. DSR is also funded by Agriculture and Food Research Initiative grant no. 2022-67015-37205/project accession no. 1028058 from the USDA National Institute of Food and Agriculture. DRP is funded by subcontract 75N93021C00014 Centers for Influenza Research and Response (CEIRR) from the National Institute of Allergy and Infectious Diseases (NIAID) and GRANT12901999, project accession no. 1022658 from the National Institute of Food and Agriculture (NIFA), U.S. Department of Agriculture. DRP receives additional support from the Georgia Research Alliance and the Caswell S. Eidson endowment funds from The University of Georgia. This study was partly supported by resources and technical expertize from the Georgia Advanced Computing Resource Center, a partnership between the University of Georgia’s Office of the Vice President for Research and the Office of the Vice President for Information Technology. A.L.B. and T.K.A. are funded by the USDA-ARS project number 5030-32000-231-000D and the NIAID, National Institutes of Health, Department of Health and Human Services, under Contract No. 75N93021C00015. The funders had no role in study design, data collection, and interpretation, or the decision to submit the work for publication. Mention of trade names or commercial products in this article is solely for the purpose of providing specific information and does not imply recommendation or endorsement by the USDA or UGA. USDA is an equal opportunity provider and employer.
Author information
Authors and Affiliations
Contributions
D.S.R. and M.C. designed the experiments. M.C., B.S., C.J.C., B.C., L.C.G., and F.C.F. performed the in vivo experiments. M.C. and B.C. processed the samples from the animal studies. M.C., B.C., and B.S. constructed the NGS libraries and sequenced the samples. B.S. performed the genetic diversity analysis and variant analysis. M.C. rescued and grew the viruses used in this study. M.C. cloned the pFastBac1 plasmids, expressed, and purified the recombinant NA proteins. M.C. performed the thermostability and NA activity assays. M.C. collected and isolated the swine tracheal cells. M.C. performed the viral growth kinetics. M.C. and B.S. performed the plaque assays and aerosol infections. M.C. performed the data analysis. M.C. and D.S.R. wrote the manuscript. D.R.P., T.K.A., and A.L.B. edited the manuscript.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Communications Biology thanks Wenjun Song and the other, anonymous, reviewers for their contribution to the peer review of this work. Primary Handling Editors: Christina Karlsson Rosenthal, Laura Rodríguez Pérez and Gavin Mason.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Cardenas, M., Seibert, B., Cowan, B. et al. Modulation of human-to-swine influenza a virus adaptation by the neuraminidase low-affinity calcium-binding pocket. Commun Biol 7, 1230 (2024). https://doi.org/10.1038/s42003-024-06928-6
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s42003-024-06928-6