Svoboda | Graniru | BBC Russia | Golosameriki | Facebook
Next Article in Journal
Sinuous Is a Claudin Required for Locust Molt in Locusta migratoria
Previous Article in Journal
PURA-Related Neurodevelopmental Disorders with Epilepsy Treated with Ketogenic Diet: A Case-Based Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Swine Influenza Viruses Isolated from 2019 to 2022 in Shandong Province, China, Exemplify the Dominant Genotype

College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai’an 271018, China
*
Author to whom correspondence should be addressed.
Genes 2024, 15(7), 849; https://doi.org/10.3390/genes15070849
Submission received: 21 May 2024 / Revised: 22 June 2024 / Accepted: 24 June 2024 / Published: 27 June 2024
(This article belongs to the Special Issue The Diversity and Evolution of the Animal Virome)

Abstract

:
Swine influenza viruses (SIVs) have been circulating in swine globally and are potential threats to human health. During the surveillance of SIVs in Shandong Province, China, from 2019 to 2022, 21 reassortant G4 genotype Eurasian avian-like (EA) H1N1 subtypes containing genes from the EA H1N1 (HA and NA), 2009 pandemic (pdm/09) H1N1 virus (PB2, PB1, PA, NP, and M), and classical swine (CS) H1N1 (NS) lineages were isolated. The analysis of the key functional amino acid sites in the isolated viruses showed that two mutation sites (190D and 225E) that preferentially bind to the human α2-6 sialic acid receptor were found in HA. In PB2, three mutation sites (271A, 590S, and 591R) that may increase mammalian fitness and a mutation site (431M) that increases pathogenicity in mice were found. A typical human signature marker that may promote infection in humans, 357K, was found in NP. The viruses could replicate efficiently in mouse lungs and turbinates, and one of the H1N1 isolates could replicate in mouse kidneys and brains without prior adaption, which indicates that the viruses potentially pose a threat to human health. Histopathological results showed that the isolated viruses caused typical bronchopneumonia and encephalitis in mice. The results indicate that G4 genotype H1N1 has potential transmissibility to humans, and surveillance should be enhanced, which could provide important information for assessing the pandemic potential of the viruses.

1. Introduction

Swine influenza is a highly contagious respiratory disease in pigs caused by influenza A virus (IAV). IAV can infect numerous animal hosts, including humans, birds, dogs, cats, pigs, horses, ferrets, minks, and seals [1]. The viral reservoir and gene pool are maintained in metapopulations of aquatic wild birds [2]. In general, IAV has high host specificity. However, genome segments can overcome the barrier through point mutations (antigenic drift) and gene reassortment (antigenic shift). The past three influenza outbreaks, including Asian influenza in 1957, Hong Kong influenza in 1968, and swine-origin H1N1 influenza in 2009, have been associated with gene reassortants derived from human and animal influenza viruses [3,4,5]. Although many IAV subtypes have been found in pigs, including H1N1, H1N2, H3N1, H3N2, H5N1, H6N6, H9N2, H10N8, and other subtypes of IAV, the H1N1, H1N2, and H3N2 subtypes are consistently isolated from swine herds globally [6,7,8,9,10,11,12,13,14]. Currently, there are four lineages circulating in China swine: the classical swine (CS) H1N1 lineage, the Eurasian avian-like (EA) H1N1 lineage, the 2009 pandemic (pdm/09) H1N1 lineage, and the North American triple-reassortant (TR) lineage [15]. Swine are considered “mixing vessels” for human and animal influenza viruses; therefore, strains with pandemic potential may be produced during mixed infections [16,17]. Improving the timely monitoring of SIVs would facilitate the assessment of the potential risks of such novel reassortant viruses to humans, in addition to their prediction and forewarning.
SIVs typically do not infect humans; however, occasionally, they can cause human infections [18,19,20,21]. The first reported case of a human infected with swine influenza was in 1958 [22]. Although the spread of SIVs between pigs and humans has been reported, so far, only pdm/09 H1N1 has been shown to be able to efficiently transmit and spread across both species. In addition, pdm/09 H1N1 has reassorted with other SIVs, generating viruses with the ability to infect humans, such as a triple-reassortant SIV, which was isolated in Fujian, China, in 2016 [23,24]. This triple-reassortant SIV has also been found in dogs in Guangxi and minks in Shandong [25,26]. In a recently published SIV surveillance study, this triple-reassortant SIV (designated as genotype 4 [G4]) was reported to be the most prevalent in pigs since 2016, and it has all the hallmarks of being highly adapted to infect humans [27].
In the present study, G4 genotypes of SIV were isolated during the epidemiological surveillance of swine herds in Shandong, China, from 2019 to 2022. Genetic characterization and pathogenicity were studied. The isolated G4 genotype strains harbored amino acid sites associated with potential transmissibility to humans, which emphasizes the importance of the timely surveillance of an emerging virus to understand its epidemiological characteristics and genetic evolution to inform the development of swine influenza prevention and control measures.

2. Materials and Methods

2.1. Ethics Statement

This study was approved by the Ethics Committee of Shandong Agricultural University (Tai’an, China). The project identification code is 2017-041, which was approved on 12 April 2017 (12/04/2017). All animal experiments were carried out in accordance with the guidelines of the Animal Care and Use Committee of Shandong Agricultural University.

2.2. Sample Collection and Treatment

A total of 7936 samples (nasal swabs = 7458; lung = 298; and trachea = 180) were collected from different intensive pig farms located in Linyi, Rizhao, Taian, Weifang, Liaocheng, Heze, Yantai, Dezhou, and Jinan in Shandong Province, China, between January 2019 and December 2022 (Table 1). The samples were collected in phosphate-buffered saline (PBS) with 2000 units/mL penicillin and 2000 units/mL streptomycin. The samples were homogenized and centrifuged at 5000× g for 5 min at 4 °C. The supernatant was then inoculated into 9-to-10-day-old specific pathogen-free (SPF) embryonated chicken eggs via the allantoic cavity (0.2 mL/embryo). After two passages in embryonated chicken eggs, the allantoic fluids were harvested and tested using a hemagglutination (HA) test with 1% chicken red blood cells, and the H1, H3, H5, and H9 subtypes of influenza viruses were confirmed by RT-PCR. Samples positive for SIV were stored at −70 °C until further use.

2.3. Nucleotide Sequencing

Viral RNA was extracted from 200 µL of virus-infected allantoic fluid using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. The RNA was reverse-transcribed into cDNA by Uni12 universal primers (5′-AGCAAAAGCAGG-3′) using the ReverTra Ace qPCR RT Kit (TOYOBO, Osaka, Japan). PCR was performed using 2× Rapid Taq Master Mix (Vazyme Biotech Co., Ltd., Nanjing, China). The complete viral genome was amplified using a total of 15 pairs of primers. All PCR products were gel-purified using the QIAquick gel extraction kit (Qiagen, Hilden, Germany). DNA sequencing and primer synthesis were carried out by Sangon Biotech (Shanghai) Co., Ltd., Shanghai, China.

2.4. Analysis of Viral Sequencing

The nucleotide sequences were edited using the SeqMan module of the DNAStar package (version 8; DNASTAR, Madison, WI, USA). The genetic similarity calculation and amino acid comparisons were performed using MegAlign in the DNAstar package (version 8; DNASTAR, Madison, WI, USA). The phylogenetic trees were constructed using MEGA 7.0 based on the neighbor-joining model with 1000 replicates. Potential glycosylation sites were identified using the NetGlyc 1.0 Server and consisted of NXS/T sites, where X denotes any amino acid, excluding proline (https://services.healthtech.dtu.dk/, accessed on 16 March 2024).

2.5. Virus Titer

Virus titration was determined using 9-to-10-day-old SPF embryonated chicken eggs and is presented as 50% egg infectious titers (EID50). Briefly, a series of 10-fold dilutions of infected allantoic fluid was prepared in sterile PBS, and each dilution (0.1 mL) was inoculated into five 9-to-10-day-old SPF embryonated chicken eggs. After 48 h of incubation at 37 °C, the allantoic fluid was harvested from each egg and tested for the presence of HA activity using the HA test. The EID50 values were calculated using the Reed–Muench method.

2.6. Infection Studies in Mice

To evaluate the pathogenicity of isolates in mice, eight-week-old SPF female BALB/c mice were divided randomly into four groups (eight mice per group). After being anesthetized lightly with dry ice, the mice were intranasally inoculated with the corresponding viruses at a dose of 106 EID50 in a 50 µL volume. Additionally, eight mice inoculated with 50 μL of PBS served as negative controls. Three mice in each group were euthanized at 3 days post-infection (dpi), and the lung, turbinate, spleen, kidney, and brain were collected for viral titration. The viral titer was determined in 9-to-10-day-old SPF embryonated chicken eggs, and the EID50 values were calculated using the Reed–Muench method. Parts of the tissue samples were fixed in 4% paraformaldehyde, embedded in paraffin, sectioned, and stained with hematoxylin and eosin. Survival and body weight were monitored for 14 days, and mice were euthanized humanely if more than 25% of the initial body weight was lost.

3. Results

3.1. Virus Isolation and Identification

A total of 7936 clinical samples collected from nine pig farms in Shandong from 2019 to 2022 were detected. Of these, 38 samples were hemagglutination-positive (0.47%), from which 21 viruses were successfully recovered. Among the 21 isolates, 15 were isolated from nasal swab samples, 4 from lung samples, and 2 from trachea samples. The isolation rates from swine in 2019, 2020, 2021, and 2022 were 38.10% (8/21), 28.57% (6/21), 19.05% (4/21), and 14.29% (3/21), respectively (Table 1). The whole genomes of all 21 isolates were sequenced and analyzed.

3.2. Phylogenetic Analysis

To investigate the genetic characteristics of the 21 isolated H1N1 viruses, all eight gene segments of the viruses were sequenced and analyzed phylogenetically. Phylogenetic analysis revealed that the hemagglutinin (HA) gene and neuraminidase (NA) gene of the 21 isolated H1N1 viruses belong to the EA H1N1 lineage (Figure 1). The HA genes of the viruses shared a 93.6–100% identity at the nucleotide level, and the NA genes shared a 93.3–100% identity. The identity of the six internal genes of the polymerase basic protein 2 (PB2), polymerase basic protein 1 (PB1), polymerase acid protein (PA), nucleoprotein (NP), matrix protein (M), and nonstructural protein (NS) genes of the 21 isolated H1N1 viruses were 94.9–100%, 95.7–100%, 95.1–100%, 94.8–100%, 95.0–100%, and 94.5–100%, respectively, at the nucleotide level. The PB2, PB1, PA, NP, and M genes of the 21 isolated H1N1 viruses exhibited the same clustering pattern and were clustered into the pdm/09 H1N1 lineage (Figure 1). The NS genes of the 21 isolated H1N1 viruses are clustered together, and they belong to the CS H1N1 lineage (Figure 1). Based on phylogenetic analyses, 21 isolated H1N1 viruses had the same genetically assembled form as the potentially pandemic G4 genotype virus.

3.3. EA H1N1 SIV Genotypes

To determine the genotypes of isolated SIVs, a total of 86 sequences of H1N1 subtype SIVs from 2008 to 2021 in Shandong Province were downloaded from the National Center for Biotechnology Information (NCBI) Influenza Virus Resource (http://www.ncbi.nlm.nih.gov/genomes/FLU/FLU.html accessed on 16 March 2024) and the Global Initiative for Sharing All Influenza Data (GISAID) database (http://www.gisaid.org), accessed on 24 December 2023. Duplicate sequences in both databases were removed by matching strain names using Bioedit v7.1.3.0 (Ibis Biosciences, Carlsbad, CA, USA). A phylogenetic analysis of the 21 isolates and 44 EA H1N1 reference strains based on the whole genome was performed (Table 2). The sequences were categorized into three genotypes (A, B, and C). All eight genomic segments of the EA H1N1 subtype isolated in Shandong from 2008 to 2013 were the avian-derived subtype, designated as genotype A. From 2014 to 2019, two distinct genotypes were characterized as triple-reassortant. Notably, EA H1N1 in genotype B was a novel triple-reassortant containing the HA, NA, and M genes of EA H1N1, the NS gene from CS H1N1, and the four remaining gene segments from pdm/09 H1N1. From 2014 to 2015, all isolated viruses belonged to genotype B. In 2016, a novel triple-reassortant SIV of genotype C was isolated. The reassortant contained the HA and NA genes derived from EA H1N1, the NS gene from CS H1N1, and the five remaining gene segments from pdm/09 H1N1. After 2016, genotype C dominated the pig population. Genotyping of the 65 SIV isolates revealed that genotype C was the most common (64.61%), followed by genotype B (26.15%) and genotype A (9.23%). All viruses isolated in Shandong Province from 2019 to 2022 belonged to genotype C.

3.4. Molecular Characterization

The molecular characteristics of all 21 isolated H1N1 viruses were analyzed based on the whole-genome sequences. Based on the deduced amino acid sequences of the HA genes, all 21 isolated H1N1 viruses possessed a single basic amino acid (PSIQSR↓G or PSIQSK↓G) in the HA cleavage site, which is characteristic of low-pathogenic avian influenza viruses (Table 3). Five potential glycosylation sites on HA (positions 14 (NST), 26 (NVT), and 277 (NCT) in HA1 and positions 484 (NGT) and 583 (NGS) in HA2) were predicted in all the isolates. For HA genes, 190D was found in 10 isolates, and 225E was found in 14 isolates, indicating that it may bind efficiently to the human α2-6 sialic acid-linked receptor [28,29]. The 225E of HA enhances the transmission of EA H1N1 SIV in guinea pigs [30]. 225E was found in the HA of 14 isolates. The 158 sites in the HA gene of all isolates were G, which may affect the antigenicity of the virus [31]. The 425M increased the replication and pathogenicity of SIV in mice [32]. All 21 isolated H1N1 viruses were not mutated at position 425. All 21 isolated H1N1 viruses had 271A and 590/591 SR in the PB2 gene, suggesting their ability to adapt to the mammalian host [33]. The 251K in the PB2 gene of EA H1N1 SIV enhances viral replication and pathogenicity in mice [34]. Among the isolates, only three strains possessed the 251K. All 21 isolates had 431M in the PB2 gene, which enhances the virulence of EA H1N1 SIV in mice [35]. The 627K and 701N affect the host range and virulence of influenza viruses [36,37,38]. None of the isolates had mutations at positions 627 and 701. The 309N enhanced the replication and pathogenicity of SIVs in mice [32]. None of the isolates exhibited mutations at this site. The 100I, 321K, 330V, and 639T enhance the pathogenicity and transmissibility of the EA H1N1 SIV [39]. None of the isolates had mutations at these positions. The amino acid substitutions associated with reduced susceptibility to NA inhibitors were not observed in all the isolated H1N1 viruses, suggesting that the isolated virus was sensitive to the antiviral drugs oseltamivir and zanamivir [40]. However, all isolates carried 31N in the M2 gene, indicating resistance to amantadine analogs [41]. The 48Q, 98K, and 99K in the NP gene enable the virus to evade human MxA restriction [42]. None of the isolates had mutations at these positions. All 21 isolated H1N1 viruses had 357K in NP, which reportedly enhances the virulence of SIVs in mice [43].

3.5. Mouse Experiments

Three isolates (A/swine/Shandong/LY059/2019 (H1N1), A/swine/Shandong/WF01/2020 (H1N1), and A/swine/Shandong/TA323/2021 (H1N1)) were selected to investigate replication and pathogenicity in mice. No significant changes in clinical signs were observed in infected mice. Mice infected with A/swine/Shandong/LY059/2019 (H1N1) had a maximum weight loss of 8.76% at 7 dpi. The virus replicated in the lungs and nasal turbinates of mice with mean titers of 5.83 log10 EID50/mL and 5.27 log10 EID50/mL, respectively, at 3 dpi. Two out of the three mice revealed detectable virus replication in the brains, and one mouse displayed detectable virus replication in the kidney, with mean titers of 2.65 and 1.8 log10 EID50, respectively. Mice infected with A/swine/Shandong/WF01/2020 (H1N1) had a maximum weight loss of 8.76% at 4 dpi. The virus replicated in the lungs and nasal turbinates of mice with mean titers of 5.83 log10 EID50/mL and 4.73 log10 EID50/mL, respectively, at 3 dpi. There was no significant weight loss in mice infected with A/swine/Shandong/TA323/2021 (H1N1) during the observation period (Figure 2A). The virus replicated in the lungs and nasal turbinates of mice with mean titers of 5.17 log10 EID50/mL and 4.47 log10 EID50/mL, respectively, at 3 dpi (Figure 2B). Histopathological analysis showed that the lungs of mice showed bronchopneumonia with changes in alveolar wall thickness, inflammatory cell infiltration, capillary congestion, and epithelial cell shedding at 3dpi (Figure 2C). Encephalitis was also observed in infected mice with vascular congestion and meningeal congestion at 3dpi (Figure 2C). The other tissues had no obvious pathological changes at 3 dpi. The results indicate that the isolated virus has low pathogenicity in mice.

4. Discussion

The EA H1N1 subtype was first isolated from pigs in Belgium in 1979, after which it spread rapidly in European and Asian countries [44]. In 2001, it was isolated in Hong Kong, China, and has been continuously present in Chinese herds [45]. Since 2005, EA H1N1 SIV has gradually become the dominant spectrum in swine herds in China [15]. After the reintroduction of pdm/09 H1N1 into swine herds, it continued to reassort with the EA H1N1 subtype and gradually replaced the pure EA H1N1 subtype, and it now dominates swine herds in China [44].
The genotypes of isolated viruses in Shandong Province were compiled, and the HA and NA genes of 65 SIVs were observed to belong to the EA H1N1 lineage, excluding 2 CS H1N1 SIVs in 2008 and 6 pdm/09 H1N1 SIVs in 2009 and 2010. In addition, no pure EA H1N1 SIVs were isolated in Shandong after 2013; however, pure EA H1N1 SIVs could still be isolated in some provinces [46]. The observations suggest that the transmission of swine influenza is geographically specific. Notably, fixed genotype B and genotype C appeared after 2014, and genotype C dominated the pig population. In the present study, 21 H1N1 subtypes, all of which belonged to genotype C, were isolated during epidemiologic surveillance in Shandong Province. The form of this genotype is identical to that of the potentially pandemic G4 EA H1N1 subtype. Previous studies have shown that this G4 EA H1N1 subtype exhibits efficient infectivity and transmission in ferret models; in addition, serologic surveillance has revealed that G4 EA H1N1 subtype infection rates can reach 10.4% and 4.4% in pig workers and the general population, respectively, indicating that the G4 EA H1N1 subtype has acquired a higher human infectivity [14]. The isolate belongs to the same genotype virus as the already reported human isolates A/Fujian-cangshan/SWL624/2016 (H1N1), A/Tianjin-baodi/1606/2018(H1N1), and A/Yunnan-Mengzi/1462/2020(H1N1) [23,24,47]. It was reported that the antibodies produced after vaccination with the current World Health Organization (WHO)-recommended vaccine strain A/Hunan/42443/2015(H1N1) did not provide effective protection against A/Yunnan-Mengzi/1462/2020(H1N1), suggesting that a new vaccine strain should be urgently constructed [9]. In addition, the G4 EA H1N1 subtype was isolated from dogs in Guangxi and from mink in Shandong, demonstrating that it has achieved cross-species transmission from pigs to dogs and mink [25,26]. The isolation of this genotype in humans, pigs, dogs, and mink-like species suggests that it has an extremely strong ability to spread across species and that this genotype is exhibiting increasing trends, making it particularly important for epidemiological surveillance and molecular evolutionary analysis.
Our analysis of the key functional amino acid substitutions of the isolated viruses revealed that HA has the 190D (10/21) and 225E (14/21) mutation sites, indicating that it can preferentially bind to the human α2-6 sialic acid receptor and cause human infection [28,29]. In addition, PB2 has the 271A and 590/591 SR (21/21) mutation sites, which may increase mammalian adaptability. PB2 has the 431M (21/21) mutation site and is capable of increasing pathogenicity in mice [35]. The NP has the 357K (21/21) mutation site, a typical human marker that may promote infection in humans [43]. The analysis of resistance sites showed that the isolated viruses were resistant to amantadine [41] and sensitive to NA inhibitors [40]. In conclusion, since mutations generated by SIV during evolution can increase adaptation, pathogenicity, and anti-influenza drug resistance in mammals, these molecular features should be monitored and studied in a timely manner.
Mice are a conventional animal model for studying SIV pathogenesis [48]. All three isolated viruses were able to replicate in the lungs and nasal turbinates of mice without prior adaptation. Among other results, virus titers indicated that the three isolated viruses replicated better in the lungs than in turbinates. The results showed that isolates could replicate better in the lower respiratory tract than in the upper respiratory tract. In addition, A/swine/Shandong/LY059/2019 (H1N1) can replicate not only in the lungs and nasal turbinates of mice but also in the brains and kidneys of mice. In our previous study, this genotypic virus did not replicate in the kidneys and brains of mice; however, our latest isolate appeared to replicate in the kidneys and brains of mice, highlighting the progressive increase in the infectivity of this genotypic strain. The exact replication mechanism in the kidney and brain remains to be further investigated. Histopathological analysis revealed changes in the lungs of mice, with alveolar wall thickening, inflammatory cell infiltration, capillary congestion, and epithelial cell detachment. In addition, vascular congestion and meningeal congestion changes were observed in the brain.

5. Conclusions

In view of the potential transmissibility of viruses from pigs to humans, the surveillance of influenza viruses transmitted between swine and human populations should be intensified. In addition, further research on virulence factors influencing influenza viruses is required to further assess their threat to public health.

Author Contributions

Conceptualization, Y.X.; data curation, Y.Z., Y.X. and Y.H.; funding acquisition, Y.X.; investigation, Y.Z., L.H., H.S. and P.Y.; methodology, Y.Z.; software, Y.Z.; supervision, Y.X. and S.L.; writing—original draft, Y.Z.; writing—review and editing, Y.X. and Y.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Major Scientific and Technological Innovation Project (MSTIP) of Shandong Province (grant no. 2023CXGC010705) and the State Key Laboratory of Veterinary Public Health and Safety (grant no. 2024SKLVPHS03).

Institutional Review Board Statement

This study was approved by the Ethics Committee of Shandong Agricultural University (Tai’an, China). The project identification code is 2017-041, which was approved on 12 April 2017 (12/04/2017). All animal experiments were carried out in accordance with the guidelines of the Animal Care and Use Committee of Shandong Agricultural University.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are included within the article.

Acknowledgments

We thank Zhang Lan (Gynecology Department of Tai’an Central Hospital) for assistance in the analysis of the gene structure.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Morens, D.M.; Taubenberger, J.K. Historical thoughts on influenza viral ecosystems, or behold a pale horse, dead dogs, failing fowl, and sick swine. Influenza Other Respir. Viruses 2010, 4, 327–337. [Google Scholar] [CrossRef] [PubMed]
  2. Nelson, M.I.; Worobey, M. Origins of the 1918 Pandemic: Revisiting the Swine “Mixing Vessel” Hypothesis. Am. J. Epidemiol. 2018, 187, 2498–2502. [Google Scholar] [CrossRef]
  3. Kawaoka, Y.; Krauss, S.; Webster, R.G. Avian-to-human transmission of the PB1 gene of influenza A viruses in the 1957 and 1968 pandemics. J. Virol. 1989, 63, 4603–4608. [Google Scholar] [CrossRef] [PubMed]
  4. Goff, P.H.; Hayashi, T.; Martínez-Gil, L.; Corr, M.; Crain, B.; Yao, S.; Cottam, H.B.; Chan, M.; Ramos, I.; Eggink, D.; et al. Synthetic Toll-like receptor 4 (TLR4) and TLR7 ligands as influenza virus vaccine adjuvants induce rapid, sustained, and broadly protective responses. J. Virol. 2015, 89, 3221–3235. [Google Scholar] [CrossRef] [PubMed]
  5. Smith, G.J.; Vijaykrishna, D.; Bahl, J.; Lycett, S.J.; Worobey, M.; Pybus, O.G.; Ma, S.K.; Cheung, C.L.; Raghwani, J.; Bhatt, S.; et al. Origins and evolutionary genomics of the 2009 swine-origin H1N1 influenza A epidemic. Nature 2009, 459, 1122–1125. [Google Scholar] [CrossRef] [PubMed]
  6. Kong, W.L.; Huang, Y.M.; Cao, N.; Qi, H.T.; Huang, L.Z.; Zhao, M.M.; Guan, S.S.; Wang, W.H.; Zhao, F.R.; Qi, W.B.; et al. Isolation and Phylogenetic Analysis of H1N1 Swine Influenza Virus from Sick Pigs in Southern China. Indian J. Virol. 2011, 22, 66–71. [Google Scholar] [CrossRef] [PubMed]
  7. Cai, M.; Huang, J.; Bu, D.; Yu, Z.; Fu, X.; Ji, C.; Zhou, P.; Zhang, G. Molecular evolution of H1N1 swine influenza in Guangdong, China, 2016–2017. Infect. Genet. Evol. 2018, 60, 103–108. [Google Scholar] [CrossRef]
  8. Jang, Y.; Seo, T.; Seo, S.H. Higher virulence of swine H1N2 influenza viruses containing avian-origin HA and 2009 pandemic PA and NP in pigs and mice. Arch. Virol. 2020, 165, 1141–1150. [Google Scholar] [CrossRef]
  9. Bowman, A.S.; Walia, R.R.; Nolting, J.M.; Vincent, A.L.; Killian, M.L.; Zentkovich, M.M.; Lorbach, J.N.; Lauterbach, S.E.; Anderson, T.K.; Davis, C.T.; et al. Influenza A(H3N2) Virus in Swine at Agricultural Fairs and Transmission to Humans, Michigan and Ohio, USA, 2016. Emerg. Infect. Dis. 2017, 23, 1551–1555. [Google Scholar] [CrossRef]
  10. Lekcharoensuk, P.; Lager, K.M.; Vemulapalli, R.; Woodruff, M.; Vincent, A.L.; Richt, J.A. Novel swine influenza virus subtype H3N1, United States. Emerg. Infect. Dis. 2006, 12, 787–794. [Google Scholar] [CrossRef]
  11. Wu, H.; Yang, F.; Lu, R.; Xu, L.; Liu, F.; Peng, X.; Wu, N. Isolation and molecular characterization of an H5N1 swine influenza virus in China in 2015. Arch. Virol. 2018, 163, 701–705. [Google Scholar] [CrossRef] [PubMed]
  12. Zhao, G.; Chen, C.; Huang, J.; Wang, Y.; Peng, D.; Liu, X. Characterisation of one H6N6 influenza virus isolated from swine in China. Res. Vet. Sci. 2013, 95, 434–436. [Google Scholar] [CrossRef] [PubMed]
  13. Xu, C.; Fan, W.; Wei, R.; Zhao, H. Isolation and identification of swine influenza recombinant A/Swine/Shandong/1/2003(H9N2) virus. Microbes Infect. 2004, 6, 919–925. [Google Scholar] [CrossRef] [PubMed]
  14. Fu, X.; Huang, Y.; Fang, B.; Liu, Y.; Cai, M.; Zhong, R.; Huang, J.; Wenbao, Q.; Tian, Y.; Zhang, G. Evidence of H10N8 influenza virus infection among swine in southern China and its infectivity and transmissibility in swine. Emerg. Microbes Infect. 2020, 9, 88–94. [Google Scholar] [CrossRef] [PubMed]
  15. Vijaykrishna, D.; Smith, G.J.; Pybus, O.G.; Zhu, H.; Bhatt, S.; Poon, L.L.; Riley, S.; Bahl, J.; Ma, S.K.; Cheung, C.L.; et al. Long-term evolution and transmission dynamics of swine influenza A virus. Nature 2011, 473, 519–522. [Google Scholar] [CrossRef] [PubMed]
  16. Wen, F.; Yu, H.; Yang, F.R.; Huang, M.; Yang, S.; Zhou, Y.J.; Li, Z.J.; Tong, G.Z. Efficacy of a high-growth reassortant H1N1 influenza virus vaccine against the classical swine H1N1 subtype influenza virus in mice and pigs. Arch. Virol. 2014, 159, 2957–2967. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, W.; Lager, K.M.; Lekcharoensuk, P.; Ulery, E.S.; Janke, B.H.; Solórzano, A.; Webby, R.J.; García-Sastre, A.; Richt, J.A. Viral reassortment and transmission after co-infection of pigs with classical H1N1 and triple-reassortant H3N2 swine influenza viruses. J. Gen. Virol. 2010, 91, 2314–2321. [Google Scholar] [CrossRef] [PubMed]
  18. Sobolev, I.; Kurskaya, O.; Leonov, S.; Kabilov, M.; Alikina, T.; Alekseev, A.; Yushkov, Y.; Saito, T.; Uchida, Y.; Mine, J.; et al. Novel reassortant of H1N1 swine influenza virus detected in pig population in Russia. Emerg. Microbes Infect. 2019, 8, 1456–1464. [Google Scholar] [CrossRef] [PubMed]
  19. Myers, K.P.; Olsen, C.W.; Gray, G.C. Cases of swine influenza in humans: A review of the literature. Clin. Infect. Dis. 2007, 44, 1084–1088. [Google Scholar] [CrossRef]
  20. Shu, B.; Garten, R.; Emery, S.; Balish, A.; Cooper, L.; Sessions, W.; Deyde, V.; Smith, C.; Berman, L.; Klimov, A.; et al. Genetic analysis and antigenic characterization of swine origin influenza viruses isolated from humans in the United States, 1990–2010. Virology 2012, 422, 151–160. [Google Scholar] [CrossRef]
  21. Gaydos, J.C.; Top, F.H., Jr.; Hodder, R.A.; Russell, P.K. Swine influenza a outbreak, Fort Dix, New Jersey, 1976. Emerg. Infect. Dis. 2006, 12, 23–28. [Google Scholar] [CrossRef] [PubMed]
  22. Krueger, W.S.; Gray, G.C. Swine influenza virus infections in man. Curr. Top. Microbiol. Immunol. 2013, 370, 201–225. [Google Scholar]
  23. Xie, J.F.; Zhang, Y.H.; Zhao, L.; Xiu, W.Q.; Chen, H.B.; Lin, Q.; Weng, Y.W.; Zheng, K.C. Emergence of Eurasian Avian-Like Swine Influenza A (H1N1) Virus from an Adult Case in Fujian Province, China. Virol. Sin. 2018, 33, 282–286. [Google Scholar] [CrossRef] [PubMed]
  24. Li, X.; Guo, L.; Liu, C.; Cheng, Y.; Kong, M.; Yang, L.; Zhuang, Z.; Liu, J.; Zou, M.; Dong, X.; et al. Human infection with a novel reassortant Eurasian-avian lineage swine H1N1 virus in northern China. Emerg. Microbes Infect. 2019, 8, 1535–1545. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, J.; Li, Z.; Cui, Y.; Yang, H.; Shan, H.; Zhang, C. Emergence of an Eurasian avian-like swine influenza A (H1N1) virus from mink in China. Vet. Microbiol. 2020, 240, 108509. [Google Scholar] [CrossRef]
  26. Chen, Y.; Trovão, N.S.; Wang, G.; Zhao, W.; He, P.; Zhou, H.; Mo, Y.; Wei, Z.; Ouyang, K.; Huang, W.; et al. Emergence and Evolution of Novel Reassortant Influenza A Viruses in Canines in Southern China. mBio 2018, 9, e00909-18. [Google Scholar] [CrossRef] [PubMed]
  27. Sun, H.; Xiao, Y.; Liu, J.; Wang, D.; Li, F.; Wang, C.; Li, C.; Zhu, J.; Song, J.; Sun, H.; et al. Prevalent Eurasian avian-like H1N1 swine influenza virus with 2009 pandemic viral genes facilitating human infection. Proc. Natl. Acad. Sci. USA 2020, 117, 17204–17210. [Google Scholar] [CrossRef] [PubMed]
  28. Glaser, L.; Stevens, J.; Zamarin, D.; Wilson, I.A.; García-Sastre, A.; Tumpey, T.M.; Basler, C.F.; Taubenberger, J.K.; Palese, P. A single amino acid substitution in 1918 influenza virus hemagglutinin changes receptor binding specificity. J. Virol. 2005, 79, 11533–11536. [Google Scholar] [CrossRef] [PubMed]
  29. Pearce, M.B.; Jayaraman, A.; Pappas, C.; Belser, J.A.; Zeng, H.; Gustin, K.M.; Maines, T.R.; Sun, X.; Raman, R.; Cox, N.J.; et al. Pathogenesis and transmission of swine origin A(H3N2)v influenza viruses in ferrets. Proc. Natl. Acad. Sci. USA 2012, 109, 3944–3949. [Google Scholar] [CrossRef]
  30. Wang, Z.; Yang, H.; Chen, Y.; Tao, S.; Liu, L.; Kong, H.; Ma, S.; Meng, F.; Suzuki, Y.; Qiao, C.; et al. A Single-Amino-Acid Substitution at Position 225 in Hemagglutinin Alters the Transmissibility of Eurasian Avian-Like H1N1 Swine Influenza Virus in Guinea Pigs. J. Virol. 2017, 91, e00800-17. [Google Scholar] [CrossRef]
  31. Wang, Z.; Chen, Y.; Chen, H.; Meng, F.; Tao, S.; Ma, S.; Qiao, C.; Chen, H.; Yang, H. A single amino acid at position 158 in haemagglutinin affects the antigenic property of Eurasian avian-like H1N1 swine influenza viruses. Transbound. Emerg. Dis. 2022, 69, e236–e243. [Google Scholar] [CrossRef] [PubMed]
  32. Hu, J.; Hu, Z.; Wei, Y.; Zhang, M.; Wang, S.; Tong, Q.; Sun, H.; Pu, J.; Liu, J.; Sun, Y. Mutations in PB2 and HA are crucial for the increased virulence and transmissibility of H1N1 swine influenza virus in mammalian models. Vet. Microbiol. 2022, 265, 109314. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, Q.; Qiao, C.; Marjuki, H.; Bawa, B.; Ma, J.; Guillossou, S.; Webby, R.J.; Richt, J.A.; Ma, W. Combination of PB2 271A and SR polymorphism at positions 590/591 is critical for viral replication and virulence of swine influenza virus in cultured cells and in vivo. J. Virol. 2012, 86, 1233–1237. [Google Scholar] [CrossRef]
  34. Cai, M.; Zhong, R.; Qin, C.; Yu, Z.; Wen, X.; Xian, J.; Chen, Y.; Cai, Y.; Yi, H.; Gong, L.; et al. The R251K Substitution in Viral Protein PB2 Increases Viral Replication and Pathogenicity of Eurasian Avian-like H1N1 Swine Influenza Viruses. Viruses 2020, 12, 52. [Google Scholar] [CrossRef]
  35. Xu, C.; Xu, B.; Wu, Y.; Yang, S.; Jia, Y.; Liang, W.; Yang, D.; He, L.; Zhu, W.; Chen, Y.; et al. A Single Amino Acid at Position 431 of the PB2 Protein Determines the Virulence of H1N1 Swine Influenza Viruses in Mice. J. Virol. 2020, 94, e01930-19. [Google Scholar] [CrossRef] [PubMed]
  36. Gong, X.Q.; Ruan, B.Y.; Liu, X.M.; Zhang, P.; Wang, X.H.; Wang, Q.; Shan, T.L.; Tong, W.; Zhou, Y.J. The PB2-K627E mutation attenuates H3N2 swine influenza virus in cultured cells and in mice. Res. Vet. Sci. 2018, 117, 54–56. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, S.; Zhu, W.; Feng, Z.; Gao, R.; Guo, J.; Li, X.; Liu, J.; Wang, D.; Shu, Y. Substitution of D701N in the PB2 protein could enhance the viral replication and pathogenicity of Eurasian avian-like H1N1 swine influenza viruses. Emerg. Microbes Infect. 2018, 7, 75. [Google Scholar] [CrossRef]
  38. Almond, J.W. A single gene determines the host range of influenza virus. Nature 1977, 270, 617–618. [Google Scholar] [CrossRef]
  39. Meng, F.; Yang, H.; Qu, Z.; Chen, Y.; Zhang, Y.; Zhang, Y.; Liu, L.; Zeng, X.; Li, C.; Kawaoka, Y.; et al. A Eurasian avian-like H1N1 swine influenza reassortant virus became pathogenic and highly transmissible due to mutations in its PA gene. Proc. Natl. Acad. Sci. USA 2022, 119, e2203919119. [Google Scholar] [CrossRef]
  40. Hussain, M.; Galvin, H.D.; Haw, T.Y.; Nutsford, A.N.; Husain, M. Drug resistance in influenza A virus: The epidemiology and management. Infect. Drug Resist. 2017, 10, 121–134. [Google Scholar] [CrossRef]
  41. He, G.; Qiao, J.; Dong, C.; He, C.; Zhao, L.; Tian, Y. Amantadine-resistance among H5N1 avian influenza viruses isolated in Northern China. Antiviral. Res. 2008, 77, 72–76. [Google Scholar] [CrossRef] [PubMed]
  42. Dornfeld, D.; Petric, P.P.; Hassan, E.; Zell, R.; Schwemmle, M. Eurasian Avian-Like Swine Influenza A Viruses Escape Human MxA Restriction through Distinct Mutations in Their Nucleoprotein. J. Virol. 2019, 93, e00997-18. [Google Scholar] [CrossRef] [PubMed]
  43. Zhu, W.; Feng, Z.; Chen, Y.; Yang, L.; Liu, J.; Li, X.; Liu, S.; Zhou, L.; Wei, H.; Gao, R.; et al. Mammalian-adaptive mutation NP-Q357K in Eurasian H1N1 Swine Influenza viruses determines the virulence phenotype in mice. Emerg. Microbes Infect. 2019, 8, 989–999. [Google Scholar] [CrossRef] [PubMed]
  44. Xu, C.; Zhang, N.; Yang, Y.; Liang, W.; Zhang, Y.; Wang, J.; Suzuki, Y.; Wu, Y.; Chen, Y.; Yang, H.; et al. Immune Escape Adaptive Mutations in Hemagglutinin Are Responsible for the Antigenic Drift of Eurasian Avian-Like H1N1 Swine Influenza Viruses. J. Virol. 2022, 96, e0097122. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, J.; Bi, Y.; Qin, K.; Fu, G.; Yang, J.; Peng, J.; Ma, G.; Liu, Q.; Pu, J.; Tian, F. Emergence of European avian influenza virus-like H1N1 swine influenza A viruses in China. J. Clin. Microbiol. 2009, 47, 2643–2646. [Google Scholar] [CrossRef]
  46. Wang, S.Y.; Wen, F.; Yu, L.X.; Wang, J.; Wang, M.Z.; Yan, J.C.; Zhou, Y.J.; Tong, W.; Shan, T.L.; Li, G.X.; et al. Potential Threats to Human Health from Eurasian Avian-Like Swine Influenza A(H1N1) Virus and Its Reassortants. Emerg. Infect. Dis. 2022, 28, 1489–1493. [Google Scholar] [CrossRef]
  47. Qin, L.; Zhang, J.J.; Chen, B.; Wang, S.F.; Yu, P.B. Genetic characteristics of the first human infection with the G4 genotype eurasian avian-like H1N1 swine influenza virus in Shaanxi Province, China. Zhonghua Yu Fang Yi Xue Za Zhi Chin. J. Prev. Med. 2023, 57, 1434–1439. [Google Scholar]
  48. Peng, X.; Wu, H.; Xu, L.; Peng, X.; Cheng, L.; Jin, C.; Xie, T.; Lu, X.; Wu, N. Molecular characterization of a novel reassortant H1N2 influenza virus containing genes from the 2009 pandemic human H1N1 virus in swine from eastern China. Virus Genes 2016, 52, 405–410. [Google Scholar] [CrossRef]
Figure 1. Phylogenetic trees of the PB2, PB1, PA, HA, NP, NA, M, and NS genes of H1N1 subtypes. The trees were generated with MEGA 7.0 using neighbor-joining analysis, and the reliabilities of the trees were assessed by bootstrap analysis with 1000 replicates. The isolates in this study are indicated by black circles.
Figure 1. Phylogenetic trees of the PB2, PB1, PA, HA, NP, NA, M, and NS genes of H1N1 subtypes. The trees were generated with MEGA 7.0 using neighbor-joining analysis, and the reliabilities of the trees were assessed by bootstrap analysis with 1000 replicates. The isolates in this study are indicated by black circles.
Genes 15 00849 g001
Figure 2. The pathogenicity of the isolates in mice. (A). Mouse body weights were monitored daily for 14 days. The values represent the average scores of overall body weight loss compared with the initial body weight ± standard deviation (SD). (B). Viral titers in lungs and nasal turbinates of the infected mice (n = 3) after 3 DPI were determined in 9-to-10-day-old SPF embryonated chicken eggs. (C). Histopathological analysis of lungs and brains. The lungs or brains of the infected mice were fixed with formalin, embedded in paraffin, stained with hematoxylin and eosin, and observed under a microscope at 200× magnification.
Figure 2. The pathogenicity of the isolates in mice. (A). Mouse body weights were monitored daily for 14 days. The values represent the average scores of overall body weight loss compared with the initial body weight ± standard deviation (SD). (B). Viral titers in lungs and nasal turbinates of the infected mice (n = 3) after 3 DPI were determined in 9-to-10-day-old SPF embryonated chicken eggs. (C). Histopathological analysis of lungs and brains. The lungs or brains of the infected mice were fixed with formalin, embedded in paraffin, stained with hematoxylin and eosin, and observed under a microscope at 200× magnification.
Genes 15 00849 g002aGenes 15 00849 g002b
Table 1. Detailed information on samples collected for SIV isolates.
Table 1. Detailed information on samples collected for SIV isolates.
Strain NameSubtypeDate CitiesHA TiterClinical Signs
A/swine/Shandong/LY059/2019H1N101/2019Linyi27Fever, coughing, sneezing
A/swine/Shandong/TA002/2019H1N102/2019Taian27NO
A/swine/Shandong/TA003/2019H1N102/2019Taian27NO
A/swine/Shandong/TA004/2019H1N102/2019Taian27NO
A/swine/Shandong/TA005/2019H1N102/2019Taian27Fever, coughing
A/swine/Shandong/TA006/2019H1N111/2019Taian26NO
A/swine/Shandong/TA008/2019H1N111/2019Taian26NO
A/swine/Shandong/LW020/2019H1N111/2019Jinan26Coughing, sneezing
A/swine/Shandong/WF01/2020H1N105/2020Weifang28Coughing, sneezing
A/swine/Shandong/DZ01/2020H1N105/2020Dezhou26NO
A/swine/Shandong/TA153/2020H1N111/2020Taian26NO
A/swine/Shandong/HZ1766/2020H1N111/2020Heze28NO
A/swine/Shandong/TA2065/2020H1N112/2020Taian28NO
A/swine/Shandong/TA3812/2020H1N112/2020Taian26NO
A/swine/Shandong/LY4407/2021H1N101/2021Linyi27NO
A/swine/Shandong/TA4500/2021H1N103/2021Taian27Fever, coughing, sneezing
A/swine/Shandong/TA323/2021H1N112/2021Taian27Fever, coughing, sneezing
A/swine/Shandong/TA6274/2021H1N112/2021Taian27NO
A/swine/Shandong/TA6303/2022H1N103/2022Taian27NO
A/swine/Shandong/TA6417/2022H1N104/2022Taian27NO
A/swine/Shandong/TA6902/2022H1N112/2022Taian26NO
NO: No obvious symptoms.
Table 2. Genotypes of H1N1 SIV in Shandong Province, China, from 2008 to 2021.
Table 2. Genotypes of H1N1 SIV in Shandong Province, China, from 2008 to 2021.
Strain NameGenotypeLineage Assigned to Gene Segments
PB2PB1PAHANPNAMNS
A/swine/Shandong/101/2008A
A/swine/Shandong/275/2009
A/swine/Shandong/327/2009
A/swine/Shandong/436/2012
A/swine/Shandong/39/2013
A/swine/Shandong/862/2013
A/swine/Shandong/S93/2014B
A/swine/Shandong/S113/2014
A/swine/Shandong/S153/2014
A/swine/Shandong/S269/2014
A/swine/Zhucheng/90/2014
A/swine/Shandong/573/2014
A/swine/Shandong/414/2014
A/swine/Shandong/807/2014
A/swine/Shandong/POS136/2015
A/swine/Shandong/POS2499/2015
A/swine/Shandong/POS2500/2015
A/swine/Shandong/POS2545/2015
A/swine/Taian/23/2017
A/swine/Shandong/123/2018
A/swine/Shandong/75/2018
A/swine/Shandong/60/2019
A/swine/Shandong/633/2019
A/swine/Shandong/1203/2016C
A/swine/Shandong/1207/2016
A/swine/Shandong/16/2016
A/swine/Shandong/36/2016
A/swine/Shandong/9/2016
A/swine/Shandong/JM78/2017
A/swine/Shandong/LY142/2017
A/swine/Laiwu/16/2017
A/swine/Shandong/0334/2017
A/swine/Shandong/0336/2017
A/swine/Shandong/0337/2017
A/swine/Shandong/10/2017
A/swine/Shandong/13/2017
A/swine/Shandong/209/2017
A/swine/Shandong/363/2017
A/swine/Shandong/451/2017
A/swine/Shandong/540/2017
A/swine/Shandong/612/2017
A/swine/Taian/95/2017
A/swine/Taian/97/2017
A/swine/China/Qingdao/2018
A/swine/Shandong/LY059/2019
A/swine/Shandong/TA002/2019
A/swine/Shandong/TA003/2019
A/swine/Shandong/TA004/2019
A/swine/Shandong/TA005/2019
A/swine/Shandong/TA006/2019
A/swine/Shandong/TA008/2019
A/swine/Shandong/LW020/2019
A/swine/Shandong/WF01/2020
A/swine/Shandong/DZ01/2020
A/swine/Shandong/TA153/2020
A/swine/Shandong/TA1766/2020
A/swine/Shandong/TA2065/2020
A/swine/Shandong/TA3812/2020
A/swine/Shandong/LY4407/2021
A/swine/Shandong/TA4500/2021
A/swine/Shandong/TA5208/2021
A/swine/Shandong/TA6274/2021
A/swine/Shandong/TA6303/2022
A/swine/Shandong/TA6417/2022
A/swine/Shandong/TA6902/2022
Genes 15 00849 i001 Genes 15 00849 i002 Genes 15 00849 i003
Table 3. Functional amino acid substitutions in the isolated viruses.
Table 3. Functional amino acid substitutions in the isolated viruses.
Gene SegmentsAmino Acid Mutations21 H1N1 Isolates
HA a
Allows efficient binding to human α2–6 sialic acid receptors190D, 225D/E [28,29]190D (10), 190V (9),190N (2); 225G (6), 225E (14), 225K (1)
Increased the transmissibility of EA H1N1 SIV in guinea pigsD225E [30]225E (14), 225G (6), 225K (1)
Influences the antigenic properties of EA H1N1 SIVE158G [31]158G (21)
Increased the replication and pathogenicity of SIV in miceL425M [32]425N (21)
PB2
A combination of 271A with the 590/591 SR polymorphism is critical for SIVs for efficient replication and adaptation in mammalsT271A-A590S-A591R [33]271A (21)-590S (21)-591R (21)
Increased viral replication and pathogenicity of the EA H1N1 SIVR251K [34]251R (18), 251K (3)
Increased the virulence of EA H1N1 SIV in mice and contributed to high polymerase activity and viral genome transcription and replicationT431M [35]431M (21)
Influences the host range and virulence of influenza viruses627K, 701N [36,37,38]627E (21), 701D (21)
Increased the replication and pathogenicity of SIVs in miceD309N [32]309D (21)
PA
Increases the pathogenicity and transmission of EA H1N1 SIV100I, 321K, 330V, 639T [39]100V (21), 321N (21), 330I (21), 639A (21)
NA b
Acquired resistance to neuraminidaseI222V, H274Y, R292K, N294S [40]222I (21), 274H (21), 292R (21), 294N (21)
M2
Resistance to amantadine-based drugs A26F, V27A, A30T/V, S31N, G34E [41]26L (21), 27V (20)/27I (1), 30A (21), 31N (21), 34G (21)
NP
Escape Human MxA Restriction48Q, 98K, 99K [42] 48K (21), 98R (21), 99R (21)
Determines the virulence phenotype in miceQ357K [43]357K (21)
a H3 numbering, which is used throughout this work; b N2 numbering, which is used throughout this work.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhao, Y.; Han, L.; Sang, H.; Liu, S.; Yang, P.; Hou, Y.; Xiao, Y. Swine Influenza Viruses Isolated from 2019 to 2022 in Shandong Province, China, Exemplify the Dominant Genotype. Genes 2024, 15, 849. https://doi.org/10.3390/genes15070849

AMA Style

Zhao Y, Han L, Sang H, Liu S, Yang P, Hou Y, Xiao Y. Swine Influenza Viruses Isolated from 2019 to 2022 in Shandong Province, China, Exemplify the Dominant Genotype. Genes. 2024; 15(7):849. https://doi.org/10.3390/genes15070849

Chicago/Turabian Style

Zhao, Yuzhong, Lebin Han, Haotian Sang, Sidang Liu, Pingping Yang, Yanmeng Hou, and Yihong Xiao. 2024. "Swine Influenza Viruses Isolated from 2019 to 2022 in Shandong Province, China, Exemplify the Dominant Genotype" Genes 15, no. 7: 849. https://doi.org/10.3390/genes15070849

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