Next Article in Journal
PRRSV Induces HMGB1 Phosphorylation at Threonine-51 Residue to Enhance Its Secretion
Next Article in Special Issue
Phylogenetic and Phylogeographic Analysis of the Highly Pathogenic H5N6 Avian Influenza Virus in China
Previous Article in Journal
A Novel In Vivo Model of Laryngeal Papillomavirus-Associated Disease Using Mus musculus Papillomavirus
Previous Article in Special Issue
Simulated Flock-Level Shedding Characteristics of Turkeys in Ten Thousand Bird Houses Infected with H7 Low Pathogenicity Avian Influenza Virus Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular Characterization and Pathogenesis of H6N6 Low Pathogenic Avian Influenza Viruses Isolated from Mallard Ducks (Anas platyrhynchos) in South Korea

1
Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea
2
Institute of Endemic Diseases, Medical Research Center, Department of Tropical Medicine and Parasitology, Seoul National University, Seoul 03080, Korea
3
Department of Tropical Medicine and Parasitology, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
4
College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2022, 14(5), 1001; https://doi.org/10.3390/v14051001
Submission received: 28 March 2022 / Revised: 3 May 2022 / Accepted: 5 May 2022 / Published: 8 May 2022
(This article belongs to the Special Issue Evolution and Epidemiology of Influenza Virus)

Abstract

:
The subtype H6N6 has been identified worldwide following the increasing frequency of avian influenza viruses (AIVs). These AIVs also have the ability to bind to human-like receptors, thereby increasing the risk of animal-human transmission. In September 2019, an H6N6 avian influenza virus—KNU2019-48 (A/Mallard (Anas platyrhynchos)/South Korea/KNU 2019-48/2019(H6N6))—was isolated from Anas platyrhynchos in South Korea. Phylogenetic analysis results revealed that the hemagglutinin (HA) gene of this strain belongs to the Korean lineage, whereas the neuraminidase (NA) and polymerase basic protein 1 (PB1) genes belong to the Chinese lineage. Outstanding internal proteins such as PB2, polymerase acidic protein, nucleoprotein, matrix protein, and non-structural protein belong to the Vietnamese lineage. Additionally, a monobasic amino acid (PRIETR↓GLF) at the HA cleavage site; non-deletion of the stalk region (residue 59–69) in the NA gene; and E627 in the PB2 gene indicate that the KNU2019-48 isolate is a typical low-pathogenic avian influenza (LPAI) virus. The nucleotide sequence similarity analysis of HA revealed that the highest homology (97.18%) of this isolate is to that of A/duck/Jiangxi/01.14 NCJD125-P/2015(H6N6), and the amino acid sequence of NA (97.38%) is closely related to that of A/duck/Fujian/10.11_FZHX1045-C/2016 (H6N6). An in vitro analysis of the KNU2019-48 virus shows a virus titer of not more than 2.8 Log10 TCID 50/mL until 72 h post-infection, whereas in the lungs, the virus is detected at 3 dpi (days post-infection). The isolated KNU2019-48 (H6N6) strain is the first reported AIV in Korea, and the H6 subtype virus has co-circulated in China, Vietnam, and Korea for half a decade. Overall, our study demonstrates that Korean H6N6 strain PB1-S375N, PA-A404S, and S409N mutations are infectious in humans and might contribute to the enhanced pathogenicity of this strain. Therefore, we emphasize the importance of continuous and intensive surveillance of the H6N6 virus not only in Korea but also worldwide.

1. Introduction

Avian influenza viruses (AIVs) belong to the Orthomyxoviridae family and are composed of eight single-stranded negative-sense RNA segments. The AIVs have been identified based on the antigenic properties of the hemagglutinin (HA) and neuraminidase (NA) glycoproteins. AIVs are categorized into 18 HA (H1–16 in wild birds and H17–18 in bats) and 11 NA (N1–9 in wild birds and N10–11 in bats) subtypes [1,2]. Furthermore, AIVs have been classified into the following two groups based on their virulence: low-pathogenic AIVs (LPAIVs) and high-pathogenic AIVs (HPAIVs). Previous studies have revealed that AIVs can have an epidemic emergence in birds, pigs, horses, and humans [3,4,5].
Downie and Laver reported in 1973 that the H6 subtype AIVs were isolated in 1965 in Massachusetts in the United States [6]. Following this discovery, the H6 subtype of AIVs has been frequently identified in isolates obtained from poultry birds, wild birds, and domestic birds around the world [7]. Over the past two decades, surveillance studies indicate that H6 subtypes have become enzootic in wild birds of southern China, mostly as H6N6 isolates. These H6 subtypes were identified by the genomic transmission of their diversity from Eurasia and North American migratory birds [8]. Simultaneously, Cheon et al. (2018) reported that many AIVs (H1N8, H1N1, H4N6, H6N2, H6N1, and H6N8) and related non-structural genes originated from a North American lineage due to intercontinental exchanges during the 2012–2017 surveillance period in South Korea [9]. However, AIV strains H6N6 and H6N2 were found co-circulating in Southern China during the surveillance periods from 2009 to 2011, and both strains showed enhanced binding to α-2,6-linked sialic acids, linked with augmented viral replication in MDCK cells [10,11,12].
Globally, H6 subtype AIVs were divided into the following two major clades: (i) The first group clade mainly consisted of H6N2 and H6N6 AIVs from Asian regions; (ii) The second group clade was mostly composed of H6N1 and H6N2 AIVs from Asian regions and other regions around the world [13]. Huang et al. found that dominant H6N2 strains were replaced with H6N6 AIVs [14]. These two AIVs strains were coexist in live bird markets in several provinces of Southern China and contain human-like receptors that are transmitted by direct contact with wild birds and guinea pigs [15].
The aim of this study is to perform a phylogenetic and mutational analysis of H6N6 AIVs isolated from mallard ducks in South Korea in 2019. Moreover, to the best of our knowledge, there are no reports related to the pathogenetic analysis of South Korean H6N6 AIVs; thus, for the first time, we elucidate the complete pathogenesis of South Korean H6N6 AIVs through in vitro and in vivo experimental infections in a mammalian model.

2. Materials and Methods

2.1. Sample Collection

During the surveillance period from January 2019 to December 2019, a total of approximately 4253 wild-bird fresh fecal samples were collected from the fields in Gyeonggi-do, South Korea (Latitude—37°23’46.8” N and Longitude—127°56’08.9” E) Figure 1. The collected samples were stored at 2–8 °C and transferred to the laboratory within 12 h for further analysis.

2.2. Virus Isolation from Feces

Fecal samples were processed according to our previously described protocol [16]. The collected fecal sample was dissolved in PBS (phosphate buffered saline; pH 7.4) supplemented with an antibiotic solution (100 U/mL penicillin and 100 mg/µL of streptomycin) (Merck, St. Louis, MO, USA) and centrifuged at 3000 rpm for 10 min at 4 °C. The supernatant was filtered through a hydrophilic polyethersulfone membrane filter having a pore size of 0.45 µm (GVS Syringe, Novatech, Kingwood, TX, USA) to remove bacteria and eukaryotic cells. The filtered supernatant samples were inoculated into the allantoic cavities of 10-day-old SPF (specific pathogen-free) ECEs (embryonated chicken eggs). Next, eggs were incubated at 37 °C for 96 h under humidified conditions, and their status was checked every day. Subsequently, eggs were chilled overnight at 4 °C, and allantoic fluids were harvested and analyzed for the confirmation of viruses based on HA (hemagglutination) activity using chicken erythrocytes.

2.3. Bird Species Identification Using the Mitochondrial Gene Cytochrome c Oxidase I (COI) as a DNA Barcode

Host bird species were determined by confirmation of a DNA barcode of a region consisting of 751 base pairs (bp) of the mitochondrial gene cytochrome c oxidase I (COXI) as previously described elsewhere [17]. The identification of the host was discovered using information from Barcode of Life Data system (BOLD; Biodiversity Institute of Ontario, University of Guelph, ON, Canada) in a combination comparison with Basic Local Alignment Search Tool for nucleotides (BLASTn; NCBI, National Institute of Health, Bethesda, MD, USA).

2.4. Extraction of Viral RNA for Sequencing

A NucleoSpin RNA kit (MACHEREY-NAGEL, Düren, Germany) was used to extract the viral RNA directly from the allantoic fluid of ECEs according to the manufacturer’s instructions. Finally, the extracted RNA was eluted in RNase-free water distributed with 20 units of RNase inhibitor and was stored at −80 °C for further analysis. Conventional real-time RT-PCR was performed to determine the presence of the influenza virus and its subtypes using total RNA following WHO (World Health Organization) guidelines [17].

2.5. Next Generation Sequencing (NGS) Analysis

NGS analysis was accompanied by GnCBIO (Dae-Jeon, Korea) on the Illumina Hiseq X platform method as previously reported [16]. Briefly, the extracted viral RNA quality was determined using an Agilent RNA 6000 Pico kit (Agilent, Santa Clara, CA, USA), and total RNA concentration was measured using a nano spectrophotometer. The cDNA library of viral RNA was constructed using a QIAseq FX single-cell RNA library kit (Qiagen, Venlo, Netherlands). Library quality was evaluated via the LightCycler qPCR system (Roche, Penzberg, Upper Bavaria, Germany) and library size was verified using TapeStation HS D5000 ScreenTape system (Agilent, Santa Clara, CA, USA). For cluster generation, the library was loaded into a flow cell where fragments were seized on a lawn of surface-bound oligos complementary to the library adapters. Using bridge amplification, each fragment was amplified into distinct clonal clusters, following which cluster creation templates were ready to be sequenced. For sequencing, data were converted into raw data for analysis. Raw sequence reads were quality-trimmed using “trim galore” (q = 20), and non-influenza virus reads were excluded using Deconseq (iden = 60). The quantity of data was corrected using a Python script up to 600,000 reads. Ephemerally, a database of only segments 4-HA, 6-NA, and 8-NS1 from the influenza virus information from NCBI were generated and aligned to those of the reference using Gsmapper (iden = 70, mL = 40). The ORF (open reading frame) was observed using the obtained consensus and adopted a result with an ORF similar to the reference. As the ORF length varied from that of the reference, sequence error was altered using Proof Read as previously described [18].

2.6. Phylogenetic Analysis and Molecular Characterization

Nucleotide blast analysis was used to identify pertinency of the viral genes. Entire reference sequences were downloaded from the NCBI (National Center for Biotechnology Information https://www.ncbi.nlm.nih.gov/ (accessed on 27 March 2022)) and GISAID (Global Initiative on Sharing All Influenza Data https://www.gisaid.org/ (accessed on 27 March 2022)) databases. Data were merged, all duplicated sequences were deleted, and phylogenetic trees were finally constructed using MEGA6.0 software (Molecular Evolutionary Genetics analysis version 6.0, Pennsylvania State University, PA, USA). Phylogenetic trees of all segments (PB2, PB1, PA, HA, NP, NA, M, and NS) of the KNU2019-48 (H6N6) viral isolate were generated by applying the maximum-likelihood with Tamura-Nei model and 1000 bootstrap replicates.

2.7. Determination of 50% Tissue Culture Infectious Dose (TCID50) and 50% Egg Infectious Dose (EID50)

ELISA (enzyme-linked immunosorbent assay) was used to measure TCID50 titers as previously reported [19], MDCK cells (ATCC, Manassas, VA, USA) were grown on 96-well flat-bottom microplates at 37 °C with 5% CO2. MDCK cells with 80–90% confluence were washed with 1× concentrated PBS and then inoculated with serial 10-fold dilutions of virus suspensions in media containing 1 µg/mL of TPCK-trypsin. Virus-infected cells were incubated at 37 °C with 5% CO2 for 72 h. Next, the TCID50 titers were determined via the Reed and Muench method [20]. To determine EID50, the allantoic cavities of 10-day-old SPF ECEs were inoculated with 100 μL serial 10-fold dilutions of the viruses, using 5 eggs for each dilution. The eggs were incubated at 37 °C for 96 h. Allantoic fluid was harvested and tested using HA assays [21], and EID50 calculation of viruses was completed using the Reed and Muench method [20].

2.8. Viral Growth Kinetics in MDCK Cells

The growth kinetics of virus isolates were evaluated in vitro. MDCK cells were infected with the viruses at an MOI (multiplicity of infection) of 0.001 in DMEM medium containing 1% antibiotic and 1 μg/mL TPCK-trypsin. Supernatants were collected every 12 hpi up to 72 hpi. ELISA was performed using anti-influenza nucleoproteins (PAN antibodies) to detect infected cells, and the virus titer (TCID50) of each supernatant was determined [22].

2.9. Animal Experiment

The pathogenic potential of the new isolate was determined in mammals using 6-week-old female BALB/c mice purchased from Orient (Seongnam, Korea) (n = 11), which were intranasally infected with 105 EID50/mL of virus. Mice were anesthetized using 1% isoflurane following the manufacturer’s instructions (Hana Pharmacy, Hwasung, Korea). The survival rate and bodyweight of mice were observed for 15 days, following which the mice were euthanized, and their lungs (n = 3 mice) were collected at 3, 6, and 15 dpi. The lung tissue was homogenized, and the TCID50 was determined to analyze the viral titers of homogenate supernatants [23]. For histopathology, the lungs of three mice were collected at 3, 6, and 15 dpi and stored in 4% formaldehyde/saline at 4 °C until further analysis. Hematoxylin and eosin (H&E) were used for histopathological examination of paraffin-embedded lung tissues mounted on glass slides. All sections were observed using a light microscope (magnification ×100). This study was approved by the Animal Ethics Committee of the Wonkwang University (WKU19-64; approved on 25 November 2019), South Korea, and all methods were conducted in accordance with relevant guidelines and regulations.

2.10. Statistics

The calculation of mean and SD (standard deviation) and Student’s t-test were performed using GraphPad Prism Software Version 5.0 (La Jolla, CA, USA). Results were presented as the mean ± SD. Statistical significance was set at p < 0.05.

3. Results

3.1. Genome Characterization of the KNU2019-48 (H6N6) Isolate

A/Mallard (Anas platyrhynchos)/South Korea/KNU 2019-48/2019(H6N6), designated as KNU2019-48 (H6N6), samples were isolated from feces of Anas platyrhynchos in South Korea on September 18, 2019. The genome sequence information of the isolate was deposited in GenBank (accession numbers MW380639 to MW380646). The GenBank accession numbers of the eight gene segments and the highest nucleotide identities from the GenBank database are shown in Table 1, with the sequence identities ranging from 96.09% to 99.21% when compared with our KNU2019-48 (H6N6) virus isolates. Surface gene HA was closely related to A/duck/Jiangxi/01.14 NCJD125-P/2015(H6N6), which originated from China, while NA was closely related to A/duck/Fujian/10.11_FZHX1045-C/2016 (H6N6) with nucleotide identities of 97.18% and 97.38%, respectively. PB2, PA, NP, M, and NS were closely related to A/duck/China/330D17/2018 (H6N6), which originated from China, with a nucleotide identity of 98.75, 99.44, 99.21, 97.59, and 98.86%, respectively. Similarly, the gene for PB1 was closely related to A/duck/Guangdong/7.20_DGCP015-C/2017 (H6N6), which originated from China, with a nucleotide similarity of 99.12%. Figure 2 shows the phylogenetic analysis of the KNU2019-48 (H6N6) strain for eight gene segments.

3.2. Hypothesis for the Reassortment Event of Each Gene Segment

Using evolutionary reassortment tracking analysis of our isolate, HA gene re-assortment prevailed from South Korea with links to China A/duck/Jiangxi/01.14 NCJD125-P/2015(H6N6) isolates in 2015. This was followed by the transmission of the HA gene from South Korea through the A/mallard/Korea/M219/2014 (H6N2) isolate in 2014. Similarly, based on the 2014–2015 report, the H6N6 strain NA gene co-circulated for a long time in China, which was later transmitted to South Korea. Next, the PB1 gene was transformed from Chinese isolates (A/duck/Guangdong/7.20_DGCP015-C/2017 (H6N6)).
The other genes (PB2, PA, NP, M, and NS) were collectively reassorted from the Chinese isolate A/duck/China/330D17/2018 (H6N6) in 2018. The backbone of these isolates originated from the Vietnamese isolates of A/Muscovy duck/Vietnam/LBM755/2014 (H5N6) in 2014. Additionally, there might have been a major H6N6 transmission from China during the migration season of 2018–2019. The locations of the putative origins of genomic components of the KNU2019-48 (H6N6) strain are shown in Figure 3.
Based on the evolutionary information, whole gene segments of our isolate KNU2019-48 (H6N6) originated from Chinese H6N6 isolates. It is possible that the Asian birds migrated through the East Asia–Australasian Flyway. Detailed information on the evolutionary re-assortment is presented in Figure 4.

3.3. Molecular Characterization of the KNU2019-48 (H6N6) Isolate

The HA cleavage site of KNU2019-48 (H6N6) contained the PRIETR↓GLF (↓denotes the cleavage site) sequence with a single basic amino acid in the HA cleavage site, which revealed a low pathogenic feature of the isolated virus. This HA receptor-binding site (RBS) suggests that H6 isolates would have a high preference toward α-2,6-linked sialic acid, which is abundant in the upper respiratory tract of avian species. We compared the HA protein of our isolate with those of the following four other isolated H6N6 strains that originated from birds and swine: (1) a low-pathogenic H6N6 isolate from domestic ducks in central China, labeled as A/duck/Hubei/10/2010 (H6N6); (2) A/swine/Guangdong/K6/2010 (H6N6) isolates, which originated from domestic ducks following whole gene reassortment; (3) isolates from poultry markets in Southern China that are a potential threat to mammals, labeled as A/duck/China/A729-2/2011(H6N6); (4) novel North American-origin avian influenza A (H6N5) virus isolated from bean goose in South Korea in 2018, which was published by our team in 2020, labeled as A/Bean goose/South Korea/KNU18-6/2018(H6N5).
A reasonable analysis of HA RBS at positions 138, 186, 190, 226, and 228 (H3 numbering) shows no mutation in amino acid residues (Table 2). The single basic residue at the HA cleavage site, with no prominent mutation at the HA RBS, showed that our KNU2019-48 (H6N6) isolates and reference strains, except for Hunan-2011, were low pathogenic H6 viruses. Moreover, the NAs of the H6N6 isolates had no amino acid deletion in the stalk (59–69) regions (Table 2). Mutations in other internal genes, also presented in Table 3, signify an enhanced viral replication efficiency along with virulence in mammals. Particularly, three human host marker mutations (PB1-S375N, PA-A404S, and S409N) were observed in KNU2019-48 (H6N6) strains, along with the mutation S375N in the PB1 gene displayed in our old H6N5 isolates from Korea, which had originated from North America. KNU2019-48 (H6N6) showed variance in certain molecular aspects, such as mutations at Q368R of PB2, K328N of PB1, S37A of PA, T223I of NA, and N30D of M1, each contributing to the potentially increased virulence of KNU2019-48 (H6N6) in mammals.

3.4. Growth Kinetics of KNU2019-48 (H6N6) Isolate in Mammalian Cell Culture

Since the molecular characterization shows reassortment via mutation, our viral isolate could increase replication efficiency; therefore, in vitro examination of the viral replication of our isolate along with other isolates was performed. To evaluate the growth kinetics of our isolate, human-origin viruses, A/California/07/2009 (H1N1) and H7N7, which are infectious to mammals, were used as controls. H1N1 replicated more efficiently in MDCK cells as compared to KNU2019-48 (H6N6) and H7N7 (Figure 5). Raw data of the TCID50 assay are shown in Figure S1.

3.5. Pathogenicity in Mice

We further examined the pathogenic potential of mice (6-week-old female BALB/c) that were intranasally infected with 105 EID50/50 µL. The H1N1 and H7N7 strains were used as the controls for comparisons. The bodyweight of the mice infected with the positive control H1N1 strain decreased from 3 dpi and was gradually regained after 10 dpi, with the lowest weight (76.15 ± 1.96%) observed at 8 dpi. Similarly, our KNU2019-48 (H6N6) isolates and the H7N7 control strain demonstrated a stable body weight trend for 15 days (Figure 6A).
All the infected mice survived the entire experimental period and showed no difference in the survival mortality between the three groups (Figure 6B). Virulence in mice lungs after 3, 6, and 15 dpi were analyzed by TCID50 assay. Based on the experimental results, control group H1N1 showed maximum virus titer in the lungs in comparison with the KNU2019-48 (H6N6) and H7N7 strains at 3 dpi (4.62 ± 0.17, 3.76 ± 0.38, 2.71 ± 0.08 log10 TCID50/mL, respectively), and these viral loads gradually decreased at 6 dpi (4.26 ± 0.44, 3.80 ± 0.63, 1.80 ± 0.05 log10 TCID50/mL, respectively) (Figure 6C). In contrast, no virus was detected in all groups at 15 dpi, indicating that these viruses were not yet well-adapted for murine infections. Further, histopathological analyses were conspicuously shown in the lungs of the virus-challenged mice. Infected lungs at 3 and 6 dpi show a condensed penetration of neutrophils into the alveolar air spaces (Figure 7).

4. Discussion

The H6N6 subtype AIV is widespread in poultry, and its host range extends to mammals such as pigs. Moreover, it has become an endemic disease in domestic animals [61]. Our isolated KNU2019-48 (H6N6) strain was co-circulated in China, Vietnam, and Korea for half a decade. These H6N6 subtype gene segments were derived from the East Asia–Australasian flyway lineages. Poultry birds may play an intermediate host role in the cross-species transmission of the influenza virus from domestic birds to humans [62]. However, this is the first isolation of H6N6 strains during our 2018 surveillance program in South Korea, and there are no detailed evidential molecular and pathological studies on H6N6 isolates from South Korea. In particular, other subtypes such as H7N3 and other H7 subtypes of AIV were pre-vailed/isolated during the surveillance period [16,63].
According to the molecular analysis of the H6N6 strain in our study, the HA cleavage site sequence PRIETR↓GLF was subtyped into a low-pathogenic virus. According to the results of phylogenetic analysis, the HA gene was closely related to A/mallard/Korea/M219/2014 (H6N2), while the genes of PB2, PA, NP, M, and NS were closer to the Chinese and Vietnamese isolates (A/duck/China/330D17/2018 (H6N6) and A/Muscovy duck/ Vietnam/LBM755/2014 (H5N6)). The PB1 gene was similar to that of the Chinese isolate, A/duck/Guangdong/7.20_DGCP015-C/2017 (H6N6), and the NA gene was similar to that of co-circulated Chinese isolates A/duck/Fujian/10.11_FZHX1045-C/2016 (H6N6) and A/chicken/Guangdong/8.30_DGCP022-O/2017(H6N6), respectively. These analyses strongly indicate that the H6 subtype virus has been frequently detected in migratory ducks in China [48,64,65].
Generally, H6 isolates receptor-binding sites were composed of HA protein residues (Q226 and G228), which preferentially bind to the α-2,3-linked sialic acid receptors in avian bird species [61]. Similarly, based on previous reports, the following HA protein residues: A137N, P186L, A193N, and G228S, were associated with human receptor-binding preference and were not found in our H6N6 isolate [7,15]. Furthermore, H6N6 isolates can be modified to replace V187D in HA responses for the binding affinity might be adapted to mammalian receptors [66]. Mutation residues E119V, H275Y, R293K, and N295S were found in the NA gene, which suggested that H6 isolates could provide insights regarding oseltamivir inhibitors. No amino acid deletions or mutations were found in the NA of KNU2019-48 (H6N6). The KNU2019-48 (H6N6) isolate carries mutations in the proteins of PB2 (Q368R, H447Q) [29,30], PB1 (D/A3V, L13P, R207K, K328N) [29,30,35], PA (F277S, C278Q, S/A515T) [43,45], NA (T223I) [53,54], M1 (N30D, V15I/T, T215A) [53,54,55], and NS1 (A/P42S) [67], which led to adaptation and increased virulence in mammals. However, the well-known mammalian adaptation markers in the protein of PB2 (E627K, D701N) were not found in our isolate [68]. Nevertheless, the KNU2019-48 (H6N6) isolate revealed human-host markers in the PB1 (S375N/T) and PA (A404S, S409N) genes [29,30,44].
Furthermore, we re-confirmed the pathogenicity of our KNU2019-48 (H6N6) isolate using in vitro experiments. We monitored the growth kinetics of KNU2019-48 (H6N6) replication in MDCK cells as follows: the virus replicated promptly and exhibited no obvious signs of illness. A maximum virus titer (2.84 log10 TCID50/mL) was achieved at 24 hpi, and there were no changes until 72 h post-infection. Similarly, our isolate strongly exhibited low pathogenicity in the lungs of mice at 3 and 6 dpi compared to the positive control H1N1, which originated from humans. Additionally, we performed body weight and survival percentage analysis. Low pathogenicity was confirmed in intranasally-infected mice; mice lungs weighed the highest at 3 dpi, whereas at 6 and 15 dpi, no variance was observed, and the lung weight was normal (Figure 6D). Although our H6N6 isolate is a low-pathogenic isolate, its movement should be monitored frequently to detect incidences of infection in humans and a potential genetic reassortment event.
In conclusion, this is the first study that broadens the knowledge of Korean H6N6 isolates from evolution to mammalian cell expressions and in vivo characterization. Continuous monitoring and molecular characterization of the H6N6 virus will be required for a better understanding of the evolutionary dynamics of the virus, which can further assist in improving control measures.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/v14051001/s1. Figure S1. Raw ELISA data of the TCID50 assay for the detection of (A) KNU2019-48 (H6N6), (B) H1N1 (CA/04/09), and (C) H7N7 growth kinetics in MDCK cells; Figure S2. Raw ELISA data of the TCID50 assay for viral load shedding in lungs after (A) 3, (B) 6, and (C) 15 days post-infection; Figure S3. Lungs from normal and infected mice at 3-, 6-, and 15-days post-infection. Scale bar: 0.5 cm; Figure S4. Lung weight at (A) 3 and (B) 15 days post-infection.

Author Contributions

Conceptualization, S.-J.Y. and H.P.; methodology, software, validation, formal analysis, K.D. and T.-T.T.T.; investigation, K.D., T.-T.T.T. and S.-Y.Y.; resources, H.-W.S.; data curation, writing—original draft preparation, K.D.; writing—review and editing, K.D., T.-T.T.T., S.-J.Y. and H.P.; visualization, K.D.; supervision, T.-T.T.T., S.-J.Y. and H.P.; project administration, S.-J.Y.; funding acquisition, H.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Priority Research Centers Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, (NRF-2015R1A6A1A03032236) and the Bio & Medical Technology Development Program of the National Research Foundation (NRF) and funded by the Korean government (MSIT) (NRF-2018M3A9H4055194).

Institutional Review Board Statement

This study was conducted according to the guidelines of the Animal Ethics Committee of Wonkwang University (WKU19-64, approval on 25 November 2019).

Informed Consent Statement

Not applicable.

Data Availability Statement

All the data presented in this study are available in this article and Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tong, S.; Li, Y.; Rivailler, P.; Conrardy, C.; Castillo, D.A.A.; Chen, L.-M.; Recuenco, S.; Ellison, J.A.; Davis, C.T.; York, I.A.; et al. A distinct lineage of influenza A virus from bats. Proc. Natl. Acad. Sci. USA 2012, 109, 4269–4274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tong, S.; Zhu, X.; Li, Y.; Shi, M.; Zhang, J.; Bourgeois, M.; Yang, H.; Chen, X.; Recuenco, S.; Gomez, J.; et al. New World Bats Harbor Diverse Influenza A Viruses. PLoS Pathog. 2013, 9, e1003657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Li, Y.; Li, M.; Tian, J.; Bai, X.; Li, Y. Genetic characteristics and pathogenicity of novel reassortant H6 viruses isolated from wild birds in China. Vet. Microbiol. 2021, 254, 108978. [Google Scholar] [CrossRef] [PubMed]
  4. Jin, Y.; Ren, H.; Teng, Y.; Hu, M.; Peng, X.; Yue, J.; Liang, L. Novel reassortment of avian influenza A(H7N9) virus with subtype H6N6 and H5N6 viruses circulating in Guangdong Province, China. J. Infect. 2017, 75, 179–182. [Google Scholar] [CrossRef]
  5. El Taweel, A.; Kandeil, A.; Barakat, A.; Alfaroq Rabiee, O.; Kayali, G.; Ali, M.A. Diversity of Astroviruses Circulating in Humans, Bats, and Wild Birds in Egypt. Viruses 2020, 12, 485. [Google Scholar] [CrossRef]
  6. Downie, J.C.; Laver, W.G. Isolation of a type A influenza virus from an Australian pelagic bird. Virology 1973, 51, 259–269. [Google Scholar] [CrossRef]
  7. Abolnik, C.; Strydom, C.; Rauff, D.L.; Wandrag, D.B.R.; Petty, D. Continuing evolution of H6N2 influenza a virus in South African chickens and the implications for diagnosis and control. BMC Vet. Res. 2019, 15, 455. [Google Scholar] [CrossRef] [Green Version]
  8. Araujo, J.; Petry, M.V.; Fabrizio, T.; Walker, D.; Ometto, T.; Thomazelli, L.M.; Scherer, A.L.; Serafini, P.P.; Neto, I.S.; Krauss, S.; et al. Migratory birds in southern Brazil are a source of multiple avian influenza virus subtypes. Influenza Other Respi. Viruses 2018, 12, 220–231. [Google Scholar] [CrossRef] [Green Version]
  9. Cheon, S.-H.; Lee, Y.-N.; Kang, S.-I.; Kye, S.-J.; Lee, E.-K.; Heo, G.-B.; Lee, M.-H.; Kim, J.-W.; Lee, K.-N.; Son, H.-M.; et al. Genetic evidence for the intercontinental movement of avian influenza viruses possessing North American-origin nonstructural gene allele B into South Korea. Infect. Genet. Evol. 2018, 66, 18–25. [Google Scholar] [CrossRef]
  10. Zou, S.; Gao, R.; Zhang, Y.; Li, X.; Chen, W.; Bai, T.; Dong, L.; Wang, D.; Shu, Y. Molecular characterization of H6 subtype influenza viruses in southern China from 2009 to 2011. Emerg. Microbes Infect. 2016, 5, 1–8. [Google Scholar] [CrossRef] [Green Version]
  11. Cui, J.; Cui, P.; Shi, J.; Fan, W.; Xing, X.; Gu, W.; Zhang, Y.; Zhang, Y.; Zeng, X.; Jiang, Y.; et al. Continued evolution of H6 avian influenza viruses isolated from farms in China between 2014 and 2018. Transbound. Emerg. Dis. 2021. [CrossRef]
  12. Wang, G.; Deng, G.; Shi, J.; Luo, W.; Zhang, G.; Zhang, Q.; Liu, L.; Jiang, Y.; Li, C.; Sriwilaijaroen, N.; et al. H6 Influenza Viruses Pose a Potential Threat to Human Health. J. Virol. 2014, 88, 3953–3964. [Google Scholar] [CrossRef] [Green Version]
  13. Everest, H.; Hill, S.; Daines, R.; Sealy, J.; James, J.; Hansen, R.; Iqbal, M. The Evolution, Spread and Global Threat of H6Nx Avian Influenza Viruses. Viruses 2020, 12, 673. [Google Scholar] [CrossRef] [PubMed]
  14. Huang, K.; Zhu, H.; Fan, X.; Wang, J.; Cheung, C.-L.; Duan, L.; Hong, W.; Liu, Y.; Li, L.; Smith, D.K.; et al. Establishment and Lineage Replacement of H6 Influenza Viruses in Domestic Ducks in Southern China. J. Virol. 2012, 86, 6075–6083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ni, F.; Kondrashkina, E.; Wang, Q. Structural and Functional Studies of Influenza Virus A/H6 Hemagglutinin. PLoS One 2015, 10, e0134576. [Google Scholar] [CrossRef] [Green Version]
  16. Trinh, T.-T.T.; Tiwari, I.; Durairaj, K.; Duong, B.T.; Nguyen, A.T.V.; Tuong, H.T.; Hoang, V.T.; Than, D.D.; Nam, S.; Yeo, S.-J.; et al. Genetic Characterization and Pathogenesis of Avian Influenza Virus H7N3 Isolated from Spot-Billed Ducks in South Korea, Early 2019. Viruses 2021, 13, 856. [Google Scholar] [CrossRef] [PubMed]
  17. Hebert, P.D.N.; Stoeckle, M.Y.; Zemlak, T.S.; Francis, C.M. Identification of Birds through DNA Barcodes. PLoS Biol. 2004, 2, e312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Lee, D.-H.; Lee, H.-J.; Lee, Y.-J.; Kang, H.-M.; Jeong, O.-M.; Kim, M.-C.; Kwon, J.-S.; Kwon, J.-H.; Kim, C.-B.; Lee, J.-B.; et al. DNA Barcoding Techniques for Avian Influenza Virus Surveillance in Migratory Bird Habitats. J. Wildl. Dis. 2010, 46, 649–654. [Google Scholar] [CrossRef] [PubMed]
  19. Yeo, S.-J.; Than, D.-D.; Park, H.-S.; Sung, H.W.; Park, H. Molecular Characterization of a Novel Avian Influenza A (H2N9) Strain Isolated from Wild Duck in Korea in 2018. Viruses 2019, 11, 1046. [Google Scholar] [CrossRef] [Green Version]
  20. Reed, L.J.; Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 1938, 27, 493–497. [Google Scholar] [CrossRef]
  21. Killian, M.L. Hemagglutination Assay for Influenza Virus. In Animal influenza virus; Humana Press: New York, NY, USA, 2014; pp. 3–9. [Google Scholar]
  22. Nguyen, N.M.; Sung, H.W.; Yun, K.-J.; Park, H.; Yeo, S.-J. Genetic Characterization of a Novel North American-Origin Avian Influenza A (H6N5) Virus Isolated from Bean Goose of South Korea in 2018. Viruses 2020, 12, 774. [Google Scholar] [CrossRef]
  23. World Health Organization. Manual for the laboratory diagnosis and virological surveillance of influenza; World Health Organization: Geneva, Switzerland, 2011; p. xii. 140p. [Google Scholar]
  24. Fan, S.; Hatta, M.; Kim, J.H.; Halfmann, P.; Imai, M.; Macken, C.A.; Le, M.Q.; Nguyen, T.; Neumann, G.; Kawaoka, Y. Novel residues in avian influenza virus PB2 protein affect virulence in mammalian hosts. Nat. Commun. 2014, 5, 5021. [Google Scholar] [CrossRef] [Green Version]
  25. Li, J.; Li, Y.; Hu, Y.; Chang, G.; Sun, W.; Yang, Y.; Kang, X.; Wu, X.; Zhu, Q. PB1-mediated virulence attenuation of H5N1 influenza virus in mice is associated with PB2. J. Gen. Virol. 2011, 92, 1435–1444. [Google Scholar] [CrossRef] [PubMed]
  26. Li, J.; Ishaq, M.; Prudence, M.; Xi, X.; Hu, T.; Liu, Q.; Guo, D. Single mutation at the amino acid position 627 of PB2 that leads to increased virulence of an H5N1 avian influenza virus during adaptation in mice can be compensated by multiple mutations at other sites of PB2. Virus Res. 2009, 144, 123–129. [Google Scholar] [CrossRef] [PubMed]
  27. Prokopyeva, E.A.; Sobolev, I.A.; Prokopyev, M.V.; Shestopalov, A.M. Adaptation of influenza A(H1N1)pdm09 virus in experimental mouse models. Infect. Genet. Evol. 2016, 39, 265–271. [Google Scholar] [CrossRef] [PubMed]
  28. Kong, H.; Ma, S.; Wang, J.; Gu, C.; Wang, Z.; Shi, J.; Deng, G.; Guan, Y.; Chen, H. Identification of Key Amino Acids in the PB2 and M1 Proteins of H7N9 Influenza Virus That Affect Its Transmission in Guinea Pigs. J. Virol. 2019, 94. [Google Scholar] [CrossRef] [Green Version]
  29. Govorkova, E.A.; Rehg, J.E.; Krauss, S.; Yen, H.-L.; Guan, Y.; Peiris, M.; Nguyen, T.D.; Hanh, T.H.; Puthavathana, P.; Long, H.T.; et al. Lethality to Ferrets of H5N1 Influenza Viruses Isolated from Humans and Poultry in 2004. J. Virol. 2005, 79, 2191–2198. [Google Scholar] [CrossRef] [Green Version]
  30. Salomon, R.; Franks, J.; Govorkova, E.A.; Ilyushina, N.A.; Yen, H.-L.; Hulse-Post, D.J.; Humberd, J.; Trichet, M.; Rehg, J.E.; Webby, R.J.; et al. The polymerase complex genes contribute to the high virulence of the human H5N1 influenza virus isolate A/Vietnam/1203/04. J. Exp. Med. 2006, 203, 689–697. [Google Scholar] [CrossRef] [Green Version]
  31. Tsurumura, T.; Qiu, H.; Yoshida, T.; Tsumori, Y.; Tsuge, H. Crystallization and preliminary X-ray diffraction studies of a surface mutant of the middle domain of PB2 from human influenza A (H1N1) virus. Acta Crystallogr. Sect. F Struct. Biol. Commun. 2014, 70, 72–75. [Google Scholar] [CrossRef] [Green Version]
  32. Li, Z.; Chen, H.; Jiao, P.; Deng, G.; Tian, G.; Li, Y.; Hoffmann, E.; Webster, R.G.; Matsuoka, Y.; Yu, K. Molecular Basis of Replication of Duck H5N1 Influenza Viruses in a Mammalian Mouse Model. J. Virol. 2005, 79, 12058–12064. [Google Scholar] [CrossRef] [Green Version]
  33. Subbarao, E.K.; London, W.; Murphy, B.R. A single amino acid in the PB2 gene of influenza A virus is a determinant of host range. J. Virol. 1993, 67, 1761–1764. [Google Scholar]
  34. Dreier, C.; Resa-Infante, P.; Thiele, S.; Stanelle-Bertram, S.; Walendy-Gnirß, K.; Speiseder, T.; Preuss, A.; Müller, Z.; Klenk, H.-D.; Stech, J.; et al. Mutations in the H7 HA and PB1 genes of avian influenza a viruses increase viral pathogenicity and contact transmission in guinea pigs. Emerg. Microbes Infect. 2019, 8, 1324–1336. [Google Scholar] [CrossRef] [Green Version]
  35. Hulse-Post, D.J.; Franks, J.; Boyd, K.; Salomon, R.; Hoffmann, E.; Yen, H.L.; Webby, R.J.; Walker, D.; Nguyen, T.D.; Webster, R.G. Molecular Changes in the Polymerase Genes (PA and PB1) Associated with High Pathogenicity of H5N1 Influenza Virus in Mallard Ducks. J. Virol. 2007, 81, 8515–8524. [Google Scholar] [CrossRef] [Green Version]
  36. Taubenberger, J.K.; Reid, A.H.; Lourens, R.M.; Wang, R.; Jin, G.; Fanning, T.G. Characterization of the 1918 influenza virus polymerase genes. Nature 2005, 437, 889–893. [Google Scholar] [CrossRef]
  37. Suttie, A.; Deng, Y.-M.; Greenhill, A.R.; Dussart, P.; Horwood, P.F.; Karlsson, E.A. Inventory of molecular markers affecting biological characteristics of avian influenza A viruses. Virus Genes 2019, 55, 739–768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Xu, C.; Hu, W.-B.; Xu, K.; He, Y.-X.; Wang, T.-Y.; Chen, Z.; Li, T.-X.; Liu, J.-H.; Buchy, P.; Sun, B. Amino acids 473V and 598P of PB1 from an avian-origin influenza A virus contribute to polymerase activity, especially in mammalian cells. J. Gen. Virol. 2012, 93, 531–540. [Google Scholar] [CrossRef] [PubMed]
  39. Feng, X.; Wang, Z.; Shi, J.; Deng, G.; Kong, H.; Tao, S.; Li, C.; Liu, L.; Guan, Y.; Chen, H. Glycine at Position 622 in PB1 Contributes to the Virulence of H5N1 Avian Influenza Virus in Mice. J. Virol. 2016, 90, 1872–1879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Shaw, M.; Cooper, L.; Xu, X.; Thompson, W.; Krauss, S.; Guan, Y.; Zhou, N.; Klimov, A.; Cox, N.; Webster, R.; et al. Molecular changes associated with the transmission of avian influenza a H5N1 and H9N2 viruses to humans. J. Med. Virol. 2002, 66, 107–114. [Google Scholar] [CrossRef]
  41. Song, J.; Xu, J.; Shi, J.; Li, Y.; Chen, H. Synergistic Effect of S224P and N383D Substitutions in the PA of H5N1 Avian Influenza Virus Contributes to Mammalian Adaptation. Sci. Rep. 2015, 5, 10510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Li, J.; Zheng, W.; Hou, L.; Chen, C.; Fan, W.; Qu, H.; Jiang, J.; Liu, J.; Gao, G.F.; Zhou, J.; et al. Differential nucleocytoplasmic shuttling of the nucleoprotein of influenza a viruses and association with host tropism. Cell. Microbiol. 2017, 19, e12692. [Google Scholar] [CrossRef] [Green Version]
  43. Leung, B.W.; Chen, H.; Brownlee, G.G. Correlation between polymerase activity and pathogenicity in two duck H5N1 influenza viruses suggests that the polymerase contributes to pathogenicity. Virology 2010, 401, 96–106. [Google Scholar] [CrossRef] [Green Version]
  44. Finkelstein, D.B.; Mukatira, S.; Mehta, P.K.; Obenauer, J.C.; Su, X.; Webster, R.G.; Naeve, C.W. Persistent Host Markers in Pandemic and H5N1 Influenza Viruses. J. Virol. 2007, 81, 10292–10299. [Google Scholar] [CrossRef] [Green Version]
  45. Yamaji, R.; Yamada, S.; Le, M.Q.; Ito, M.; Sakai-Tagawa, Y.; Kawaoka, Y. Mammalian Adaptive Mutations of the PA Protein of Highly Pathogenic Avian H5N1 Influenza Virus. J. Virol. 2015, 89, 4117–4125. [Google Scholar] [CrossRef] [Green Version]
  46. Su, Y.; Yang, H.-Y.; Zhang, B.-J.; Jia, H.-L.; Tien, P. Analysis of a point mutation in H5N1 avian influenza virus hemagglutinin in relation to virus entry into live mammalian cells. Arch. Virol. 2008, 153, 2253–2261. [Google Scholar] [CrossRef]
  47. Gao, Y.; Zhang, Y.; Shinya, K.; Deng, G.; Jiang, Y.; Li, Z.; Guan, Y.; Tian, G.; Li, Y.; Shi, J.; et al. Identification of Amino Acids in HA and PB2 Critical for the Transmission of H5N1 Avian Influenza Viruses in a Mammalian Host. PLOS Pathog. 2009, 5, e1000709. [Google Scholar] [CrossRef] [Green Version]
  48. Tan, L.; Su, S.; Smith, D.K.; He, S.; Zheng, Y.; Shao, Z.; Ma, J.; Zhu, H.; Zhang, G. A Combination of HA and PA Mutations Enhances Virulence in a Mouse-Adapted H6N6 Influenza A Virus. J. Virol. 2014, 88, 14116–14125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Zhu, W.; Zou, X.; Zhou, J.; Tang, J.; Shu, Y. Residues 41V and/or 210D in the NP protein enhance polymerase activities and potential replication of novel influenza (H7N9) viruses at low temperature. Virol. J. 2015, 12, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Liu, Q.; Chen, H.; Huang, J.; Chen, Y.; Gu, M.; Wang, X.; Hu, S.; Liu, X.; Liu, X. A nonpathogenic duck-origin H9N2 influenza A virus adapts to high pathogenicity in mice. Arch. Virol. 2014, 159, 2243–2252. [Google Scholar] [CrossRef]
  51. Ma, S.; Zhang, B.; Shi, J.; Yin, X.; Wang, G.; Cui, P.; Liu, L.; Deng, G.; Jiang, Y.; Li, C.; et al. Amino Acid Mutations A286V and T437M in the Nucleoprotein Attenuate H7N9 Viruses in Mice. J. Virol. 2020, 94. [Google Scholar] [CrossRef] [Green Version]
  52. Zhao, Y.; Yu, Z.; Liu, L.; Wang, T.; Sun, W.; Wang, C.; Xia, Z.; Gao, Y.; Zhou, B.; Qian, J.; et al. Adaptive amino acid substitutions enhance the virulence of a novel human H7N9 influenza virus in mice. Vet. Microbiol. 2016, 187, 8–14. [Google Scholar] [CrossRef]
  53. Chen, H.; Bright, R.A.; Subbarao, K.; Smith, C.; Cox, N.J.; Katz, J.M.; Matsuoka, Y. Polygenic virulence factors involved in pathogenesis of 1997 Hong Kong H5N1 influenza viruses in mice. Virus Res. 2007, 128, 159–163. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, M.S.; Deng, M.C.; Lin, Y.J.; Chang, C.Y.; Shieh, H.K.; Shiau, J.Z.; Huang, C.C. Characterization of an H5N1 avian influenza virus from Taiwan. Vet. Microbiol. 2007, 124, 193–201. [Google Scholar] [CrossRef]
  55. Fan, S.; Deng, G.; Song, J.; Tian, G.; Suo, Y.; Jiang, Y.; Guan, Y.; Bu, Z.; Kawaoka, Y.; Chen, H. Two amino acid residues in the matrix protein M1 contribute to the virulence difference of H5N1 avian influenza viruses in mice. Virology 2009, 384, 28–32. [Google Scholar] [CrossRef] [Green Version]
  56. Jiao, P.; Tian, G.; Li, Y.; Deng, G.; Jiang, Y.; Liu, C.; Liu, W.; Bu, Z.; Kawaoka, Y.; Chen, H. A Single-Amino-Acid Substitution in the NS1 Protein Changes the Pathogenicity of H5N1 Avian Influenza Viruses in Mice. J. Virol. 2008, 82, 1146–1154. [Google Scholar] [CrossRef] [Green Version]
  57. Zheng, W.; Cao, S.; Chen, C.; Li, J.; Zhang, S.; Jiang, J.; Niu, Y.; Fan, W.; Li, Y.; Bi, Y.; et al. Threonine 80 phosphorylation of non-structural protein 1 regulates the replication of influenza A virus by reducing the binding affinity with RIG-I. Cell. Microbiol. 2017, 19, e12643. [Google Scholar] [CrossRef] [Green Version]
  58. Min, J.-Y.; Li, S.; Sen, G.C.; Krug, R.M. A site on the influenza A virus NS1 protein mediates both inhibition of PKR activation and temporal regulation of viral RNA synthesis. Virology 2007, 363, 236–243. [Google Scholar] [CrossRef] [Green Version]
  59. Li, Z.; Jiang, Y.; Jiao, P.; Wang, A.; Zhao, F.; Tian, G.; Wang, X.; Yu, K.; Bu, Z.; Chen, H. The NS1 Gene Contributes to the Virulence of H5N1 Avian Influenza Viruses. J. Virol. 2006, 80, 11115–11123. [Google Scholar] [CrossRef] [Green Version]
  60. Imai, H.; Shinya, K.; Takano, R.; Kiso, M.; Muramoto, Y.; Sakabe, S.; Murakami, S.; Ito, M.; Yamada, S.; Le, M. thi Q.; et al. The HA and NS Genes of Human H5N1 Influenza A Virus Contribute to High Virulence in Ferrets. PLoS Pathog. 2010, 6, e1001106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Lin, W.; Cui, H.; Teng, Q.; Li, L.; Shi, Y.; Li, X.; Yang, J.; Liu, Q.; Deng, J.; Li, Z. Evolution and pathogenicity of H6 avian influenza viruses isolated from Southern China during 2011 to 2017 in mice and chickens. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef] [PubMed]
  62. Tsai, S.-K.; Shih, C.-H.; Chang, H.-W.; Teng, K.-H.; Hsu, W.-E.; Lin, H.-J.; Lin, H.-Y.; Huang, C.-H.; Chen, H.-W.; Wang, L.-C. Replication of a Dog-Origin H6N1 Influenza Virus in Cell Culture and Mice. Viruses 2020, 12, 704. [Google Scholar] [CrossRef] [PubMed]
  63. Duong, B.T.; Bal, J.; Sung, H.W.; Yeo, S.-J.; Park, H. Molecular Analysis of the Avian H7 Influenza Viruses Circulating in South Korea during 2018–2019: Evolutionary Significance and Associated Zoonotic Threats. Viruses 2021, 13, 2260. [Google Scholar] [CrossRef] [PubMed]
  64. Kang, Y.; Liu, L.; Feng, M.; Yuan, R.; Huang, C.; Tan, Y.; Gao, P.; Xiang, D.; Zhao, X.; Li, Y.; et al. Highly pathogenic H5N6 influenza A viruses recovered from wild birds in Guangdong, southern China, 2014–2015. Sci. Rep. 2017, 7, 44410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bi, Y.; Li, J.; Li, S.; Fu, G.; Jin, T.; Zhang, C.; Yang, Y.; Ma, Z.; Tian, W.; Xiao, S.; et al. Dominant subtype switch in avian influenza viruses during 2016–2019 in China. Nat. Commun 2020, 11, 5909. [Google Scholar] [CrossRef]
  66. Zhang, G.; Kong, W.; Qi, W.; Long, L.-P.; Cao, Z.; Huang, L.; Qi, H.; Cao, N.; Wang, W.; Zhao, F.; et al. Identification of an H6N6 swine influenza virus in southern China. Infect. Genet. Evol. 2011, 11, 1174–1177. [Google Scholar] [CrossRef] [Green Version]
  67. Shanmuganatham, K.K.; Jones, J.C.; Marathe, B.M.; Feeroz, M.M.; Jones-Engel, L.; Walker, D.; Turner, J.; Rabiul Alam, S.M.; Kamrul Hasan, M.; Akhtar, S.; et al. The replication of Bangladeshi H9N2 avian influenza viruses carrying genes from H7N3 in mammals. Emerg. Microbes Infect. 2016, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
  68. Miura, H.; Ozeki, Y.; Omatsu, T.; Katayama, Y.; Imai, K.; Mizutani, T.; Ogawa, H.; Takeda, Y. The Single E627K Amino Acid Substitution in PB2 Enhances the Pathogenicity of Wild-Bird-Origin H6N6 Subtype Avian Influenza Virus in Mice. Austin J. Infect. Dis. 2020, 7, 1041. [Google Scholar]
Figure 1. Google Earth image for sample colleting location with the information of latitude and longitude.
Figure 1. Google Earth image for sample colleting location with the information of latitude and longitude.
Viruses 14 01001 g001
Figure 2. (AH) Phylogenetic analysis of the KNU2019-48 (H6N6) strain for eight gene segments. (A) PB2; (B) PB1; (C) PA; (D) HA; (E) NP; (F) NA; (G) M; (H) NS. (PB—polymerase basic protein; NP—nucleoprotein; HA—hemagglutinin; PA—polymerase acidic protein; NA—neuraminidase; M—matrix protein; NS—non-structural protein).
Figure 2. (AH) Phylogenetic analysis of the KNU2019-48 (H6N6) strain for eight gene segments. (A) PB2; (B) PB1; (C) PA; (D) HA; (E) NP; (F) NA; (G) M; (H) NS. (PB—polymerase basic protein; NP—nucleoprotein; HA—hemagglutinin; PA—polymerase acidic protein; NA—neuraminidase; M—matrix protein; NS—non-structural protein).
Viruses 14 01001 g002aViruses 14 01001 g002b
Figure 3. Locations of the putative origins of genomic components of the KNU2019-48 (H6N6) strain.
Figure 3. Locations of the putative origins of genomic components of the KNU2019-48 (H6N6) strain.
Viruses 14 01001 g003
Figure 4. Original reassortment events of the novel avian influenza isolate KNU2019-48 (H6N6).
Figure 4. Original reassortment events of the novel avian influenza isolate KNU2019-48 (H6N6).
Viruses 14 01001 g004
Figure 5. Growth kinetics of KNU2019-48 (H6N6) replication in MDCK cells. Three kinds of viruses were infected into MDCK cells at a multiplicity of infection (MOI) of 0.001. The cell supernatants were collected at different time-points (12, 24, 36, 48, 60, 72, and 84 hpi). The virus titer concentration in cell culture supernatant was determined by an enzyme-linked immunosorbent assay (ELISA) using anti-influenza nucleoprotein (NP) to detect infected cells, and TCID50 was determined in MDCK cells. Data are represented as mean ± SD and calculated from three replicates. **, p < 0.01; ***, p < 0.001.
Figure 5. Growth kinetics of KNU2019-48 (H6N6) replication in MDCK cells. Three kinds of viruses were infected into MDCK cells at a multiplicity of infection (MOI) of 0.001. The cell supernatants were collected at different time-points (12, 24, 36, 48, 60, 72, and 84 hpi). The virus titer concentration in cell culture supernatant was determined by an enzyme-linked immunosorbent assay (ELISA) using anti-influenza nucleoprotein (NP) to detect infected cells, and TCID50 was determined in MDCK cells. Data are represented as mean ± SD and calculated from three replicates. **, p < 0.01; ***, p < 0.001.
Viruses 14 01001 g005
Figure 6. In vivo pathogenicity of the KNU2019-48 (H6N6) virus isolate. BALB/c mice were intranasally infected with 105 EID50 concentrations of the virus per mouse. H1N1 and H7N7 virus isolates were used as control. Mean changes in (A) body weight, (B) survival rates, (C) virus titers in the lung, and (D) lung weight were noted. Body weights are presented as percentages of the original weight (n = 5). **, p < 0.01; ***, p < 0.001.
Figure 6. In vivo pathogenicity of the KNU2019-48 (H6N6) virus isolate. BALB/c mice were intranasally infected with 105 EID50 concentrations of the virus per mouse. H1N1 and H7N7 virus isolates were used as control. Mean changes in (A) body weight, (B) survival rates, (C) virus titers in the lung, and (D) lung weight were noted. Body weights are presented as percentages of the original weight (n = 5). **, p < 0.01; ***, p < 0.001.
Viruses 14 01001 g006
Figure 7. Histology of lung inflammation determined by hematoxylin and eosin (H&E) staining. For each isolate, BALB/c mice were intranasally infected with 105 EID50 concentrations of the virus per mouse. The uninfected control (normal); KNU2019-48 (H6N6)-, H1N1 (CA/04/09)-, and H7N7-infected mouse lungs were collected and stained with H&E at days 3, 6, and 15 post infection (dpi) (scale bar, 100 µm; original magnification ×100).
Figure 7. Histology of lung inflammation determined by hematoxylin and eosin (H&E) staining. For each isolate, BALB/c mice were intranasally infected with 105 EID50 concentrations of the virus per mouse. The uninfected control (normal); KNU2019-48 (H6N6)-, H1N1 (CA/04/09)-, and H7N7-infected mouse lungs were collected and stained with H&E at days 3, 6, and 15 post infection (dpi) (scale bar, 100 µm; original magnification ×100).
Viruses 14 01001 g007
Table 1. Virus strains obtained from the GenBank database with highest nucleotide identities when compared with the KNU2019-48 (H6N6) isolate in this study.
Table 1. Virus strains obtained from the GenBank database with highest nucleotide identities when compared with the KNU2019-48 (H6N6) isolate in this study.
GeneGenBank IDReference Strain
Accession ID
OriginPer Ident (%)
PB2MW380639EPI_ISL_501514A/duck/China/330D17/2018 (H6N6)98.75 (2341/2328)
EPI_ISL_285466A/duck/Fujian/SD086/2017 (H6N6)98.29 (2280/2328)
EPI666098A/duck/Guangdong/02.11_DGCPLB005-P/2015 (H6N6)97.16 (2335/2328)
PB1MW380640EPI_ISL_707456A/duck/Guangdong/7.20_DGCP015-C/2017 (H6N6)99.12 (2274/2304)
MW104102A/chicken/Guangdong/7.20_DGCP050-O/2017(mixed)99.03 (2274/2304)
EPI_ISL_698000A/chicken/Guangdong/7.20_DGCP050-O/2017 (H9N2)99.03 (2274/2304)
PAMW380641EPI_ISL_501514A/duck/China/330D17/2018 (H6N6)99.44 (2233/2151)
EPI_ISL_285466A/duck/Fujian/SD086/2017 (H6N6)98.14 (2151/2151)
EPI_ISL_76327A/duck/Shantou/2472/2005 (H6N2)96.09 (2151/2151)
HAMW380642EPI_ISL_199312A/duck/Jiangxi/01.14 NCJD125-P/2015(H6N6)97.18 (1740/1701)
MH130170A/mallard/Korea/M219/2014 (H6N2)96.47 (1726/1701)
EPI_ISL_219853A/Environment/Hunan/02483/2014 (H6N6)98 (1701/1701)
NPMW380643EPI_ISL_501514A/duck/China/330D17/2018 (H6N6)99.21 (1565/1527)
EPI_ISL_696839A/duck/Guizhou/10.28_ZYLJJ001-C/2018 (H6N6)98.33 (1497/1527)
MW098939A/duck/Guangdong/7.20_DGCP030-C/2017(mixed)97.33 (1497/1527)
NAMW380644EPI_ISL_696964A/duck/Fujian/10.11_FZHX1045-C/2016 (H6N6)97.38 (1412/1465)
EPI666988A/duck/Guangxi/04.10_JX019/2015 (H6N6)96.10 (1412/1465)
MW100376A/chicken/Inner_mongolia/12.02_EEDSWSQ002-C/2018 (H6N6)95.47 (1413/1465)
MMW380645MN088783A/duck/China/330D17/2018 (H6N6)97.59 (1027/979)
MW101275A/duck/Fujian/11.26_FZHX0181-C/2018(mixed)99.18 (976/979)
LC028304A/muscovy duck/Vietnam/LBM755/2014(H5N6)99.18 (976/979)
NSMW380646MN088790A/duck/China/330D17/2018(H6N6)98.86 (890/889)
MW101859A/duck/Guizhou/10.28_ZYLJJ001-C/2018(H6N6)97.75 (844/889)
CY109470A/duck/Shantou/17490/2006(H6N2)97.16 (844/889)
Table 2. Comparison between the HA receptor-binding sites and NA of the avian influenza H6N6 virus and other host-pathogenic H6 viruses.
Table 2. Comparison between the HA receptor-binding sites and NA of the avian influenza H6N6 virus and other host-pathogenic H6 viruses.
Virus StrainsHA Receptor-Binding Residues (H3 Numbering)NA
Cleavage Sites
340-348
A138SP186LE190VQ226LG228SStalk Region DeletionE119VH275YR293KN295S
KNU2019-48 (H6N6)PRIETR↓GLFAPEQGNOEHRN
H10/2010 (H6N6)PQIETR↓GLFAPEQGNOEHRN
K6/2010 (H6N6)PQIETR↓GLFSIEQSNOEHRN
A729-2/2011 (H6N6)PQIETR↓GLFAPEQGYES (59–69)EHRN
KNU18-6/2018 (H6N5)PQIETR↓GLFAPEQGNOEHRN
KNU2019-48 (H6N6): A/Mallard (Anas platyrhynchos)/South Korea/KNU 2019-48/2019(H6N6), H10/2010 (H6N6): A/duck/Hubei/10/2010 (H6N6), K6/2010 (H6N6): A/swine/Guangdong/K6/2010 (H6N6), A729-2/2011 (H6N6): A/duck/China/A729-2/2011(H6N6), KNU18-6/2018 (H6N5): A/Bean goose/South Korea/KNU18-6/2018(H6N5). ↓ cleavage site.
Table 3. Summary of data obtained from the mutational analysis of eight genes from AIVs of multiple avian species with the KNU2019-48 (H6N6) isolate.
Table 3. Summary of data obtained from the mutational analysis of eight genes from AIVs of multiple avian species with the KNU2019-48 (H6N6) isolate.
Viral ProteinAmino AcidKNU2019-48
(H6N6)
H10/2010
(H6N6)
K6/2010
(H6N6)
A72-2/2011
(H6N6)
KNU18-6/2018
(H6N5)
PhenotypeReference
PB2K147T,
M147L
IIIII-[24]
T63I (with PB1 M677T)IIIIIPathogenic in mice[25]
L89VVVVVVEnhanced polymerase activity; Increased virulence in mice[26]
K251RRRRRRIncreased virulence in mice[27]
I292VIIIIIIncrease the polymerase activity of
H7N9 viruses in both avian and human cells and facilitate the transmission
[28]
G309DDDDDDEnhanced polymerase activity; Increased virulence in mice[26]
T339KKKKKKEnhanced polymerase activity; Increased virulence in mice
Q368RRRRRRIncreased polymerase activity; Increased virulence in mammals[29,30]
H447QQQQQQIncreased polymerase activity; Increased virulence in mammals
I471T (with PB2 P453H)TTTTTChange the surface electrostatic potential drastically[31]
R477GGGGGGEnhanced polymerase activity; Increased virulence in mice[26]
I495VVVVVVEnhanced polymerase activity; Increased virulence in mice
A676TTTTTTEnhanced polymerase active; Increased virulence in mice
E627KEEEEEMammalian adaptation marker[32,33]
D701NDDDDDMammalian adaptation marker
PB1D/A3VVVVVVIncreased polymerase activity; Increased virulence in mammals[29,30]
L13PPP-PPIncreased polymerase activity; Increased virulence in mammals, Mammalian host marker[34]
R207KKKKKKIncreased polymerase activity in mammalian cells[35]
K328NNNNNNIncreased polymerase activity; Increased virulence in mammals[29,30]
S375N/TNNNNNIncreased polymerase activity; Increased virulence in mammals, Human host marker
H436YYYYYYIncreased polymerase activity and virulence in mallards, ferrets, and mice[36]
A469T
(with NS1
N205K;
NEP T48N)
TTTTTConferred in contact transmissibility in guinea pigs[35]
L473VVVVVVIncreased polymerase activity and replication efficiency
V652AAAAAAIncreased virulence in mice[27]
M677T (with PB2 T63I)IIIVIPathogenic in mice[25]
V598PLLLLLDecreased polymerase activity and replication efficiency in mammalian cells[37,38]
D622GGGGGGIncreased polymerase activity and virulence in mice[39]
PAN383DDDDDDIncreased polymerase activity in mammalian and avian cell lines[40,41]
S37AAAAAASignificantly increased viral growth and polymerase activity in mammalian cells[42]
H266RRRRRRIncreased polymerase activity; Increased virulence in mammals and birds[43]
F277SSSSSSAdapt to mammalian hosts
C278QQQQQQAdapt to mammalian hosts
I357KTTTTTIncreased polymerase activity; Increased virulence in mammals and birds
N383D (with S224P)DDDDDEnhanced the pathogenicity and viral replication of H5N1 virus in mice[40,41]
A404SSSSSAHuman host marker[44]
S409NNNNNSEnhanced Transmission; Human host marker
S/A515TTTTTTIncreased polymerase activity; Increased virulence in mammals and birds[43,45]
L653PPPPPPAdapt to mammalian hosts[43]
HAV110AAAAAAHost specificity shift to Enhance binding of HA to human-type SAα2,6Gal receptor[46]
T160AEGSAEIncreased binding to human-type influenza receptor[47]
T/E173G/D/VDDDDT Increased virus binding to α-2,6-linked sialic acid[22,48]
NPV41IIIIIIMight contribute to viral transmissibility[49]
V105MMIMMMContribute to the increased virulence of the H9N2[50]
D210EEEEEEMight contribute to viral transmissibility [49]
F253IIIIIIResults in attenuated pathogenicity of the virus in mice[42]
A286VAAAAAAffect the
pathogenicity of the virus in mice
[51]
I353VVVVVVIncreased virulence in mice[27]
T437MTTTTTAffect the
pathogenicity of the virus in mice
[51]
NAM26IVIII Increased virulence in mice [52]
T223IIIII Increased virulence in mammals[53,54]
M1N30DDDDDDIncreased virulence in mammals[55]
V15I/TIIIIIIncreased virulence in mammals[53,54]
A166VVVVVVContribute to the increased virulence of the H9N2.[49]
T215AAAAAAIncreased virulence in mammals[55]
NS1A/P42SSSSSSIncreased virulence in mammals; Antagonism of IFN induction[56]
T80ENTTTTReduced influenza virus replication through controlling RIG-I-mediated IFN production and vRNP activity[57]
T/D/V/R/A127NDNNNNIncreased virulence in mammals[58]
V149AAAAAAPathogenicity in mice; Antagonism of IFN induction[59]
NS2T47A (with NS1 N200S)EEEEEDecreased IFN antagonism[60]
M51I (with NS1 G205R)RRRRRDecreased IFN antagonism
(“-“. no phenotypes were found).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Durairaj, K.; Trinh, T.-T.T.; Yun, S.-Y.; Yeo, S.-J.; Sung, H.-W.; Park, H. Molecular Characterization and Pathogenesis of H6N6 Low Pathogenic Avian Influenza Viruses Isolated from Mallard Ducks (Anas platyrhynchos) in South Korea. Viruses 2022, 14, 1001. https://doi.org/10.3390/v14051001

AMA Style

Durairaj K, Trinh T-TT, Yun S-Y, Yeo S-J, Sung H-W, Park H. Molecular Characterization and Pathogenesis of H6N6 Low Pathogenic Avian Influenza Viruses Isolated from Mallard Ducks (Anas platyrhynchos) in South Korea. Viruses. 2022; 14(5):1001. https://doi.org/10.3390/v14051001

Chicago/Turabian Style

Durairaj, Kaliannan, Thuy-Tien Thi Trinh, Su-Yeon Yun, Seon-Ju Yeo, Haan-Woo Sung, and Hyun Park. 2022. "Molecular Characterization and Pathogenesis of H6N6 Low Pathogenic Avian Influenza Viruses Isolated from Mallard Ducks (Anas platyrhynchos) in South Korea" Viruses 14, no. 5: 1001. https://doi.org/10.3390/v14051001

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop