1
|
Elhusseiny MH, Elsayed MM, Mady WH, Mahana O, Bakry NR, Abdelaziz O, Arafa AS, Shahein MA, Eid S, Naguib MM. Genetic features of avian influenza (A/H5N8) clade 2.3.4.4b isolated from quail in Egypt. Virus Res 2024; 350:199482. [PMID: 39396573 DOI: 10.1016/j.virusres.2024.199482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Several genotypes of the highly pathogenic avian influenza (HPAI) virus H5N8 subtype within clade 2.3.4.4b continue to circulate in different species of domestic birds across Egypt. It is believed that quail contribute to virus replication and adaptation to other gallinaceous poultry species and humans. This study provides genetic characterization of the full genome of HPAI H5N8 isolated from quail in Egypt. The virus was isolated from a commercial quail farm associated with respiratory signs. To characterize the genetic features of the detected virus, gene sequencing via Sanger technology and phylogenetic analysis were performed. The results revealed high nucleotide identity with the HPAI H5N8 virus from Egypt, which has multiple basic amino acid motifs PLREKRRKR/GLF at the hemagglutinin (HA) cleavage site. Phylogenetic analysis of the eight gene segments revealed that the quail isolate is grouped with HPAI H5N8 viruses of clade 2.3.4.4b and closely related to the most recent circulating H5N8 viruses in Egypt. Whole-genome characterization revealed amino acid preferences for avian receptors with few mutations, indicating their affinity for human-like receptors and increased virulence in mammals, such as S123P, S133A, T156A and A263T in the HA gene. In addition, the sequencing results revealed a lack of markers associated with influenza antiviral resistance in the neuraminidase and matrix-2 coding proteins. The results of the present study support the spread of HPAIV H5N8 to species other than chickens in Egypt. Therefore, continuous surveillance of AIV in different bird species in Egypt followed by full genomic characterization is needed for better virus control and prevention.
Collapse
Affiliation(s)
- Mohamed H Elhusseiny
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Moataz M Elsayed
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Wesam H Mady
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Osama Mahana
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Neveen R Bakry
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Ola Abdelaziz
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Abdel-Sattar Arafa
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | | | - Samah Eid
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Mahmoud M Naguib
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt; Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK.
| |
Collapse
|
2
|
Li Q, Jiang L, Wang Y, Liu X, Wang B, Shan Z, Wang YH, Wang Y, Chen H, Li C. HACD3 Prevents PB1 from Autophagic Degradation to Facilitate the Replication of Influenza A Virus. Viruses 2024; 16:702. [PMID: 38793585 PMCID: PMC11126133 DOI: 10.3390/v16050702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Influenza A virus (IAV) continues to pose serious threats to the global animal industry and public health security. Identification of critical host factors engaged in the life cycle of IAV and elucidation of the underlying mechanisms of their action are particularly important for the discovery of potential new targets for the development of anti-influenza drugs. Herein, we identified Hydroxyacyl-CoA Dehydratase 3 (HACD3) as a new host factor that supports the replication of IAV. Downregulating the expression of HACD3 reduced the level of viral PB1 protein in IAV-infected cells and in cells that were transiently transfected to express PB1. Silencing HACD3 expression had no effect on the level of PB1 mRNA but could promote the lysosome-mediated autophagic degradation of PB1 protein. Further investigation revealed that HACD3 interacted with PB1 and selective autophagic receptor SQSTM1/p62, and HACD3 competed with SQSTM1/p62 for the interaction with PB1, which prevented PB1 from SQSTM1/p62-mediated autophagic degradation. Collectively, these findings establish that HACD3 plays a positive regulatory role in IAV replication by stabilizing the viral PB1 protein.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Q.L.); (L.J.); (Y.W.); (X.L.); (B.W.); (Z.S.); (Y.-H.W.); (Y.W.)
| | - Chengjun Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Q.L.); (L.J.); (Y.W.); (X.L.); (B.W.); (Z.S.); (Y.-H.W.); (Y.W.)
| |
Collapse
|
3
|
Bedair NM, Sakr MA, Mourad A, Eissa N, Mostafa A, Khamiss O. Molecular characterization of the whole genome of H9N2 avian influenza virus isolated from Egyptian poultry farms. Arch Virol 2024; 169:99. [PMID: 38625394 PMCID: PMC11021324 DOI: 10.1007/s00705-024-06018-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/13/2024] [Indexed: 04/17/2024]
Abstract
H9N2 avian influenza viruses (AIVs) affect both poultry and humans on a global level, and they are especially prevalent in Egypt. In this study, we sequenced the entire genome of AIV H9N2 isolated from chickens in Egypt in 2021, using next-generation sequencing (NGS) technology. Phylogenetic analysis of the resulting sequences showed that the studied strain was generally monophyletic and grouped within the G1 sublineage of the Eurasian lineage. Four segments (polymerase basic 2 [PB2], polymerase basic 1 [PB1], polymerase acidic [PA], and non-structural [NS]) were related to Egyptian genotype II, while the nucleoprotein (NP), neuraminidase (NA), matrix (M), and haemagglutinin (HA) segments were related to Egyptian genotype I. Molecular analysis revealed that HA protein contained amino acid residues (191H and 234L) that suggested a predilection for attaching to human-like receptors. The antigenic sites of HA had two nonsynonymous mutations: V194I at antigenic site A and M40K at antigenic site B. Furthermore, the R403W and S372A mutations, which have been observed in H3N2 and H2N2 strains that caused human pandemics, were found in the NA protein of the detected strain. The internal proteins contained virulence markers: 504V in the PB2 protein, 622G, 436Y, 207K, and 677T in the PB1 protein, 127V, 550L, and 672L in PA protein, and 64F and 69P in the M protein. These results show that the detected strain had undergone intrasubtype reassortment. Furthermore, it contains changes in the viral proteins that make it more likely to be virulent, raising a question about the tendency of AIV H9N2 to become highly pathogenic in the future for both poultry and humans.
Collapse
Affiliation(s)
- Nahed M Bedair
- Molecular Diagnostics and Therapeutics Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City (USC), Sadat, Egypt
| | - Moustafa A Sakr
- Molecular Diagnostics and Therapeutics Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City (USC), Sadat, Egypt.
| | - Ahmed Mourad
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, University of Sadat City, Sadat, Egypt
| | - Nourhan Eissa
- Department of Animal Hygiene and Zoonoses, Faculty of Veterinary Medicine, University of Sadat City, Sadat, Egypt
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, 12622, Dokki, Giza, Egypt
| | - Omaima Khamiss
- Animal Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City (USC), Sadat, Egypt
| |
Collapse
|
4
|
Kutkat O, Gomaa M, Aboulhoda BE, Moatasim Y, El Taweel A, Kamel MN, El Sayes M, Elkhrsawy A, AbdAllah H, Kandeil A, McKenzie PP, Webby RJ, Ali MA, Kayali G, El-Shesheny R. Genetic and virological characteristics of a reassortant avian influenza A H6N1 virus isolated from wild birds at a live-bird market in Egypt. Arch Virol 2024; 169:95. [PMID: 38594485 DOI: 10.1007/s00705-024-06022-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/13/2024] [Indexed: 04/11/2024]
Abstract
The first detection of a human infection with avian influenza A/H6N1 virus in Taiwan in 2013 has raised concerns about this virus. During our routine surveillance of avian influenza viruses (AIVs) in live-bird markets in Egypt, an H6N1 virus was isolated from a garganey duck and was characterized. Phylogenetic analysis indicated that the Egyptian H6N1 strain A/Garganey/Egypt/20869C/2022(H6N1) has a unique genomic constellation, with gene segments inherited from different subtypes (H5N1, H3N8, H7N3, H6N1, and H10N1) that have been detected previously in AIVs from Egypt and some Eurasian countries. We examined the replication of kinetics of this virus in different mammalian cell lines (A549, MDCK, and Vero cells) and compared its pathogenicity to that of the ancestral H6N1 virus A/Quail/HK/421/2002(H6N1). The Egyptian H6N1 virus replicated efficiently in C57BL/6 mice without prior adaptation and grew faster and reached higher titers than in A549 cells than the ancestral strain. These results show that reassortant H6 AIVs might pose a potential threat to human health and highlight the need to continue surveillance of H6 AIVs circulating in nature.
Collapse
Affiliation(s)
- Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Mokhtar Gomaa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Mina Nabil Kamel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Mohamed El Sayes
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Amany Elkhrsawy
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Hend AbdAllah
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Pamela P McKenzie
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Mohamed Ahmed Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | | | - Rabeh El-Shesheny
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt.
| |
Collapse
|
5
|
Flores RA, Cammayo-Fletcher PLT, Nguyen BT, Villavicencio AGM, Lee SY, Son Y, Kim JH, Park KI, Yoo WG, Jin YB, Min W, Kim WH. Genetic Characterization and Phylogeographic Analysis of the First H13N6 Avian Influenza Virus Isolated from Vega Gull in South Korea. Viruses 2024; 16:285. [PMID: 38400060 PMCID: PMC10891532 DOI: 10.3390/v16020285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Avian influenza virus (AIV) is a pathogen with zoonotic and pandemic potential. Migratory birds are natural reservoirs of all known subtypes of AIVs, except for H17N10 and H18N11, and they have been implicated in previous highly pathogenic avian influenza outbreaks worldwide. This study identified and characterized the first isolate of the H13N6 subtype from a Vega gull (Larus vegae mongolicus) in South Korea. The amino acid sequence of hemagglutinin gene showed a low pathogenic AIV subtype and various amino acid substitutions were found in the sequence compared to the reference sequence and known H13 isolates. High sequence homology with other H13N6 isolates was found in HA, NA, PB1, and PA genes, but not for PB2, NP, M, and NS genes. Interestingly, various point amino acid mutations were found on all gene segments, and some are linked to an increased binding to human-type receptors, resistance to antivirals, and virulence. Evolutionary and phylogenetic analyses showed that all gene segments are gull-adapted, with a phylogeographic origin of mostly Eurasian, except for PB2, PA, and M. Findings from this study support the evidence that reassortment of AIVs continuously occurs in nature, and migratory birds are vital in the intercontinental spread of avian influenza viruses.
Collapse
Affiliation(s)
- Rochelle A. Flores
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Paula Leona T. Cammayo-Fletcher
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Binh T. Nguyen
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Andrea Gail M. Villavicencio
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Seung Yun Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Yongwoo Son
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Jae-Hoon Kim
- National Park Research Institute, Korean National Park Service, Wonju 26441, Gangwon, Republic of Korea;
| | - Kwang Il Park
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Won Gi Yoo
- Department of Parasitology and Tropical Medicine, College of Medicine, Gyeongsang National University, Jinju 52727, Gyeongnam, Republic of Korea;
| | - Yeung Bae Jin
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Wongi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| | - Woo H. Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Gyeongnam, Republic of Korea; (R.A.F.); (P.L.T.C.-F.); (B.T.N.); (A.G.M.V.); (S.Y.L.); (Y.S.); (K.I.P.); (Y.B.J.); (W.M.)
| |
Collapse
|
6
|
Li J, Takeda M, Imahatakenaka M, Ikeda M. Identification of dihydroorotate dehydrogenase inhibitor, vidofludimus, as a potent and novel inhibitor for influenza virus. J Med Virol 2024; 96:e29372. [PMID: 38235544 DOI: 10.1002/jmv.29372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 12/14/2023] [Accepted: 12/25/2023] [Indexed: 01/19/2024]
Abstract
Influenza A virus (IAV) infection causes respiratory disease. Recently, infection of IAV H5N1 among mammals are reported in farmed mink. Therefore, to discover antivirals against IAV, we screened a compound library by using the RNA-dependent RNA polymerase (RdRp) assay system derived from H5N1 IAV including a drug-resistant PA mutant (I38T) and a viral polymerase activity enhancing PB2 mutant (T271A). Upon screening, we found vidofludimus can be served as a potential inhibitor for IAV. Vidofludimus an orally active inhibitor for dihydroorotate dehydrogenase (DHODH), a key enzyme for the cellular de novo pyrimidine biosynthesis pathway. We found that vidofludimus exerted antiviral activity against wild-type and drug-resistant mutant IAV, with effective concentrations (EC50 ) of 2.10 and 2.11 μM, respectively. The anti-IAV activity of vidofludimus was canceled by the treatment of uridine or cytidine through pyrimidine salvage synthesis pathway, or orotic acid through pyrimidine de novo synthesis pathway. This indicated that the main target of vidofludimus is DHODH in IAV RdRp expressing cells. We also produced recombinant seasonal IAV H1N1 virion and influenza B virus (IBV) RdRp assay system and confirmed vidofludimus also carried highly antiviral activity against seasonal IAV and IBV. Vidofludimus is a candidate drug for the future threat of IAV H5N1 infection among humans as well as seasonal influenza virus infection.
Collapse
Affiliation(s)
- Jiazhou Li
- Division of Biological Information Technology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Midori Takeda
- Division of Biological Information Technology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Mikiko Imahatakenaka
- Division of Biological Information Technology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Masanori Ikeda
- Division of Biological Information Technology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
7
|
Wang Y, Wang M, Zhang H, Zhao C, Zhang Y, Shen J, Sun X, Xu H, Xie Y, Gao X, Cui P, Chu D, Li Y, Liu W, Peng P, Deng G, Guo J, Li X. Prevalence, evolution, replication and transmission of H3N8 avian influenza viruses isolated from migratory birds in eastern China from 2017 to 2021. Emerg Microbes Infect 2023; 12:2184178. [PMID: 36913241 PMCID: PMC10013397 DOI: 10.1080/22221751.2023.2184178] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The continued evolution and emergence of novel influenza viruses in wild and domestic animals poses an increasing public health risk. Two human cases of H3N8 avian influenza virus infection in China in 2022 have caused public concern regarding the risk of transmission between birds and humans. However, the prevalence of H3N8 avian influenza viruses in their natural reservoirs and their biological characteristics are largely unknown. To elucidate the potential threat of H3N8 viruses, we analyzed five years of surveillance data obtained from an important wetland region in eastern China and evaluated the evolutionary and biological characteristics of 21 H3N8 viruses isolated from 15,899 migratory bird samples between 2017 and 2021. Genetic and phylogenetic analyses showed that the H3N8 viruses circulating in migratory birds and ducks have evolved into different branches and have undergone complicated reassortment with viruses in waterfowl. The 21 viruses belonged to 12 genotypes, and some strains induced body weight loss and pneumonia in mice. All the tested H3N8 viruses preferentially bind to avian-type receptors, although they have acquired the ability to bind human-type receptors. Infection studies in ducks, chickens and pigeons demonstrated that the currently circulating H3N8 viruses in migratory birds have a high possibility of infecting domestic waterfowl and a low possibility of infecting chickens and pigeons. Our findings imply that circulating H3N8 viruses in migratory birds continue to evolve and pose a high infection risk in domestic ducks. These results further emphasize the importance of avian influenza surveillance at the wild bird and poultry interface.
Collapse
Affiliation(s)
- Yanwen Wang
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Mengjing Wang
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Hong Zhang
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Conghui Zhao
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin, People's Republic of China
| | - Yaping Zhang
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin, People's Republic of China
| | - Jinyan Shen
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Xiaohong Sun
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Hongke Xu
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Yujiao Xie
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Xinxin Gao
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Pengfei Cui
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin, People's Republic of China
| | - Dong Chu
- Biological Disaster Control and Prevention Center, National Forestry and Grassland Administration, Shenyang, People's Republic of China
| | - Yubao Li
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Wenqiang Liu
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Peng Peng
- Biological Disaster Control and Prevention Center, National Forestry and Grassland Administration, Shenyang, People's Republic of China
| | - Guohua Deng
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin, People's Republic of China
| | - Jing Guo
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| | - Xuyong Li
- College of Agronomy, Liaocheng University, Liaocheng, People's Republic of China
| |
Collapse
|
8
|
Alasiri A, Soltane R, Hegazy A, Khalil AM, Mahmoud SH, Khalil AA, Martinez-Sobrido L, Mostafa A. Vaccination and Antiviral Treatment against Avian Influenza H5Nx Viruses: A Harbinger of Virus Control or Evolution. Vaccines (Basel) 2023; 11:1628. [PMID: 38005960 PMCID: PMC10675773 DOI: 10.3390/vaccines11111628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Despite the panzootic nature of emergent highly pathogenic avian influenza H5Nx viruses in wild migratory birds and domestic poultry, only a limited number of human infections with H5Nx viruses have been identified since its emergence in 1996. Few countries with endemic avian influenza viruses (AIVs) have implemented vaccination as a control strategy, while most of the countries have adopted a culling strategy for the infected flocks. To date, China and Egypt are the two major sites where vaccination has been adopted to control avian influenza H5Nx infections, especially with the widespread circulation of clade 2.3.4.4b H5N1 viruses. This virus is currently circulating among birds and poultry, with occasional spillovers to mammals, including humans. Herein, we will discuss the history of AIVs in Egypt as one of the hotspots for infections and the improper implementation of prophylactic and therapeutic control strategies, leading to continuous flock outbreaks with remarkable virus evolution scenarios. Along with current pre-pandemic preparedness efforts, comprehensive surveillance of H5Nx viruses in wild birds, domestic poultry, and mammals, including humans, in endemic areas is critical to explore the public health risk of the newly emerging immune-evasive or drug-resistant H5Nx variants.
Collapse
Affiliation(s)
- Ahlam Alasiri
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.A.); (R.S.)
| | - Raya Soltane
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.A.); (R.S.)
| | - Akram Hegazy
- Department of Agricultural Microbiology, Faculty of Agriculture, Cairo University, Giza District, Giza 12613, Egypt;
| | - Ahmed Magdy Khalil
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
- Department of Zoonotic Diseases, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Sara H. Mahmoud
- Center of Scientific Excellence for Influenza Viruses, National Research Center, Giza 12622, Egypt;
| | - Ahmed A. Khalil
- Veterinary Sera and Vaccines Research Institute (VSVRI), Agriculture Research Center (ARC), Cairo 11435, Egypt;
| | | | - Ahmed Mostafa
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
- Center of Scientific Excellence for Influenza Viruses, National Research Center, Giza 12622, Egypt;
| |
Collapse
|
9
|
Lee SH, Lee J, Noh JY, Jeong JH, Kim JB, Kwon JH, Youk S, Song CS, Lee DH. Age is a determinant factor in the susceptibility of domestic ducks to H5 clade 2.3.2.1c and 2.3.4.4e high pathogenicity avian influenza viruses. Front Vet Sci 2023; 10:1207289. [PMID: 37546334 PMCID: PMC10400362 DOI: 10.3389/fvets.2023.1207289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
High pathogenicity avian influenza (HPAI) is a viral disease with devastating consequences for the poultry industry worldwide. Domestic ducks are a major source of HPAI viruses in many Eurasian countries. The infectivity and pathogenicity of HPAI viruses in ducks vary depending on host and viral factors. To assess the factors influencing the infectivity and pathogenicity of HPAI viruses in ducks, we compared the pathobiology of two HPAI viruses (H5N1 clade 2.3.2.1c and H5N6 clade 2.3.4.4e) in 5- and 25-week-old ducks. Both HPAI viruses caused mortality in a dose-dependent manner (104, 106, and 108 EID50) in young ducks. By contrast, adult ducks were infected but exhibited no mortality due to either virus. Viral excretion was higher in young ducks than in adults, regardless of the HPAI strain. These findings demonstrate the age-dependent mortality of clade 2.3.2.1c and clade 2.3.4.4e H5 HPAI viruses in ducks.
Collapse
Affiliation(s)
- Sun-Hak Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jiho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jin-Yong Noh
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- KHAV Co., Ltd., Seoul, Republic of Korea
| | - Jei-Hyun Jeong
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- KHAV Co., Ltd., Seoul, Republic of Korea
| | - Jun-Beom Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- KHAV Co., Ltd., Seoul, Republic of Korea
| | - Jung-Hoon Kwon
- Laboratory of Veterinary Microbiology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sungsu Youk
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju-si, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- KHAV Co., Ltd., Seoul, Republic of Korea
| | - Dong-Hun Lee
- Wildlife Health Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Fernández-Díaz M, Villanueva-Pérez D, Tataje-Lavanda L, Montalvan-Avalos A, Isasi-Rivas G, Lulo-Vargas M, Fernández-Sánchez M. Detection and Genomic Characterization of an Avian Influenza Virus Subtype H5N1 (Clade 2.3.4.4b) Strain Isolated from a Pelican in Peru. Microbiol Resour Announc 2023; 12:e0019923. [PMID: 37125942 PMCID: PMC10281143 DOI: 10.1128/mra.00199-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 05/02/2023] Open
Abstract
Surveillance helps us identify and monitor strains with zoonotic potential. A tracheal swab from a pelican on a Peruvian beach was H5N1 positive (clade 2.3.4.4b) using Oxford Nanopore's MinION platform. The near-complete genome sequence of strain VFAR-140 will aid us in understanding avian influenza epidemiology and spread.
Collapse
Affiliation(s)
- Manolo Fernández-Díaz
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
| | - Doris Villanueva-Pérez
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
| | - Luis Tataje-Lavanda
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Peru
| | - Angela Montalvan-Avalos
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
| | - Gisela Isasi-Rivas
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
| | - Milagros Lulo-Vargas
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
| | - Manolo Fernández-Sánchez
- Laboratorios de Investigación y Desarrollo, Farmacológicos Veterinarios S.A.C. (FARVET SAC), Chincha Alta, Ica, Peru
| |
Collapse
|
11
|
Petherbridge G, Gadzhiev AA, Shestopalov АМ, Alekseev AY, Sharshov KA, Daudova MG. An early warning system for highly pathogenic viruses borne by waterbird species and related dynamics of climate change in the Caspian Sea region: Outlines of a concept. SOUTH OF RUSSIA: ECOLOGY, DEVELOPMENT 2022. [DOI: 10.18470/1992-1098-2022-2-233-263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aim. Formulation of the outlines of the concept of ViEW (Viral Early Warning) which is intended as a long term system of multidisciplinary transboundary cooperation between specialist institutions of all five Caspian region states to research, regularly monitor and share data about the generation, transmission and epidemiology of avian‐borne pathogens and their vectors in the region, and the ways climate change may affect these processes.Material and Methods. The concept is based on the multidisciplinary experience of the authors in researching the processes incorporated in the ViEW concept and on an in‐depth survey of the literature involved.Results. The outlines of the ViEW concept are presented in this study for review and comment by interested parties and stakeholders.Conclusion. Review of activities and opinions of specialists and organizations with remits relating to the development, establishment and maintenance of ViEW, indicates that such a system is a necessity for global animal and human health because of the role that the Caspian region plays in the mass migration of species of waterbird known as vectors for avian influenza and the already evident impacts of climate change on their phenologies. Waterbirds frequenting the Caspian Sea littorals and their habitats together constitute a major potential global hotspot or High Risk region for the generation and transmission of highly pathogenic avian influenza viruses and other dangerous zoonotic diseases.
Collapse
Affiliation(s)
| | | | - А. М. Shestopalov
- Research Institute of Virology, Federal Research Centre of Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences
| | - A. Yu. Alekseev
- Research Institute of Virology, Federal Research Centre of Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences
| | - K. A. Sharshov
- Research Institute of Virology, Federal Research Centre of Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences
| | | |
Collapse
|
12
|
El-Shesheny R, Moatasim Y, Mahmoud SH, Song Y, El Taweel A, Gomaa M, Kamel MN, Sayes ME, Kandeil A, Lam TTY, McKenzie PP, Webby RJ, Kayali G, Ali MA. Highly Pathogenic Avian Influenza A(H5N1) Virus Clade 2.3.4.4b in Wild Birds and Live Bird Markets, Egypt. Pathogens 2022; 12:36. [PMID: 36678384 PMCID: PMC9866256 DOI: 10.3390/pathogens12010036] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/15/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
Clade 2.3.4.4 H5Nx influenza viruses have further diversified into several subclades. Sub-clade 2.3.4.4b H5N1 viruses have been widely circulating in wild birds and detected in Europe, Africa, Asia, and North America since October 2020. In this study, we report the first detection of highly pathogenic avian influenza H5N1 clade 2.3.4.4b viruses in wild birds and domestic ducks from live bird markets in Egypt. Phylogenetic analysis revealed that the Egyptian H5N1 virus retained the genomic composition of Eurasian strains. Mutations in the viral proteins associated with zoonotic potential and pathogenicity were detected in Egyptian isolates. Egypt is considered a hot spot for the evolution of the influenza virus, so active surveillance of avian influenza viruses in Egypt is warranted.
Collapse
Affiliation(s)
- Rabeh El-Shesheny
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Sara H. Mahmoud
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Yi Song
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited, Hong Kong SAR, China
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Mokhtar Gomaa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Mina Nabil Kamel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Mohamed El Sayes
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Tommy T. Y. Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited, Hong Kong SAR, China
- Centre for Immunology & Infection Limited, Hong Kong SAR, China
| | - Pamela P. McKenzie
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ghazi Kayali
- Human Link, Dubai 3O-01-BA380, United Arab Emirates
| | - Mohamed Ahmed Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| |
Collapse
|
13
|
Yang Q, Xue X, Zhang Z, Wu MJ, Ji J, Wang W, Yin H, Li S, Dai H, Duan B, Liu Q, Song J. Clade 2.3.4.4b H5N8 Subtype Avian Influenza Viruses Were Identified from the Common Crane Wintering in Yunnan Province, China. Viruses 2022; 15:38. [PMID: 36680078 PMCID: PMC9863098 DOI: 10.3390/v15010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
The seasonal migration of wild aquatic birds plays a critical role in the maintenance, transmission, and incursion of the avian influenza virus (AIV). AIV surveillance was performed during 2020-2021 in two national nature reserves with abundant wild bird resources in Yunnan, China. Four H5N8 AIVs isolates from the common crane were identified by next-generation sequencing. Phylogenetic analysis demonstrated that all eight gene segments of these H5N8 AIVs belonged to clade 2.3.4.4b high-pathogenic AIV (HPAIV) and shared high nucleotide sequence similarity with the strains isolated in Hubei, China, and Siberia, Russia, in 2020-2021. The H5N8 HPAIVs from common cranes were characterized by both human and avian dual-receptor specificity in the hemagglutinin (HA) protein. Moreover, possessing the substitutions contributes to overcoming transmission barriers of mammalian hosts in polymerase basic 2 (PB2), polymerase basic protein 1 (PB1), and polymerase acid (PA), and exhibiting the long stalk in the neck region of the neuraminidase (NA) protein contributes to adaptation in wild birds. Monitoring AIVs in migratory birds, at stopover sites and in their primary habitats, i.e., breeding or wintering grounds, could provide insight into potential zoonosis caused by AIVs.
Collapse
Affiliation(s)
- Qinhong Yang
- College of Life Sciences, Southwest Forestry University, 300 Bailong Road, Kunming 650024, China
| | - Xiaoyan Xue
- College of Life Sciences, Southwest Forestry University, 300 Bailong Road, Kunming 650024, China
| | - Zhenxing Zhang
- Yunnan Tropical and Subtropical Animal Virus Diseases Laboratory, Yunnan Academy of Animal Husbandry and Veterinary, 6 Qinglongshan, Kunming 650224, China
| | - Ming J. Wu
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Jia Ji
- College of Life Sciences, Southwest Forestry University, 300 Bailong Road, Kunming 650024, China
| | - Wei Wang
- College of Life Sciences, Southwest Forestry University, 300 Bailong Road, Kunming 650024, China
| | - Hongbin Yin
- Animal Disease Inspection and Supervision Institution of Yunnan Province, 118 Gulou Road, Kunming 650051, China
| | - Suhua Li
- College of Life Sciences, Southwest Forestry University, 300 Bailong Road, Kunming 650024, China
| | - Hongyang Dai
- The Management Bureau of Huize Black—Necked Crane National Nature Reserve, 744 Tongbao Road, Qujing 654200, China
| | - Bofang Duan
- Yunnan Center for Animal Disease Control and Prevention, 95 Jinhei Road, Kunming 650034, China
| | - Qiang Liu
- College of Life Sciences, Southwest Forestry University, 300 Bailong Road, Kunming 650024, China
| | - Jianling Song
- Yunnan Tropical and Subtropical Animal Virus Diseases Laboratory, Yunnan Academy of Animal Husbandry and Veterinary, 6 Qinglongshan, Kunming 650224, China
| |
Collapse
|
14
|
Genetic Analysis of a Novel H16N3 Virus Isolated from a Migratory Gull in China in 2021 and Animal Studies of Infection. Microbiol Spectr 2022; 10:e0248422. [PMID: 36314919 PMCID: PMC9769943 DOI: 10.1128/spectrum.02484-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
H16 avian influenza viruses mainly circulate in wild migratory gulls worldwide, and the infection risks in poultry and mammals remain largely unknown. In this study, we isolated a novel H16N3 virus from migratory gulls in eastern China in 2021. Genetic analysis indicated that the H16N3 virus originated from the H16 and H13 viruses that circulated in wild birds. This H16N3 virus has not adapted to replicate in chickens, ducks, or mice, although it can be transmitted between inoculated and contacted birds. The circulation of H16Nx viruses in the Northern Hemisphere indicates that we should strengthen active surveillance to monitor their prevalence and evolution in migratory gulls and their introduction into other migratory and domestic waterfowl. IMPORTANCE Migratory wild birds are natural reservoirs of H16 viruses and play a key role in the global prevalence of these viruses. Here, we found that H16 viruses predominantly circulate in migratory gulls and that the gull H16N3 virus cannot replicate efficiently in chickens, ducks, or mice without prior adaptation. These findings contribute to our understanding of the ecology, evolution, and biological properties of H16 viruses and will guide avian influenza surveillance in birds.
Collapse
|
15
|
Liu T, Huang Y, Xie S, Xu L, Chen J, Qi W, Liao M, Jia W. A Characterization and an Evolutionary and a Pathogenicity Analysis of Reassortment H3N2 Avian Influenza Virus in South China in 2019-2020. Viruses 2022; 14:v14112574. [PMID: 36423183 PMCID: PMC9692712 DOI: 10.3390/v14112574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Seasonal H3N2 influenza virus has always been a potential threat to public health. The reassortment of the human and avian H3N2 influenza viruses has resulted in major influenza outbreaks, which have seriously damaged human life and health. To assess the possible threat of the H3N2 avian influenza virus to human health, we performed whole-genome sequencing and genetic evolution analyses on 10 H3N2 field strains isolated from different hosts and regions in 2019-2020 and selected representative strains for pathogenicity tests on mice. According to the results, the internal gene cassettes of nine strains had not only undergone reassortment with the H1, H2, H4, H6, and H7 subtypes, which circulate in poultry and mammals, but also with H10N8, which circulates in wild birds in the natural environment. Three reassorted strains were found to be pathogenic to mice, of these one strain harboring MP from H10N8 showed a stronger virulence in mice. This study indicates that reassorted H3N2 AIVs may cross the host barrier to infect mammals and humans, thereby, necessitating persistent surveillance of H3N2 AIVs.
Collapse
Affiliation(s)
- Tengfei Liu
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yuhao Huang
- Center for Animal Disease Control and Prevention, Dongguan 523128, China
| | - Shumin Xie
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lingyu Xu
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Junhong Chen
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wenbao Qi
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Ming Liao
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Weixin Jia
- National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: ; Tel.: +86-13826409229
| |
Collapse
|
16
|
Zhang L, Liu K, Su Q, Chen X, Wang X, Li Q, Wang W, Mao X, Xu J, Zhou X, Xu Q, Zhou L, Liu X, Zhang P. Clinical features of the first critical case of acute encephalitis caused by avian influenza A (H5N6) virus. Emerg Microbes Infect 2022; 11:2437-2446. [PMID: 36093829 PMCID: PMC9621215 DOI: 10.1080/22221751.2022.2122584] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Highly pathogenic avian influenza viruses (HPAIV), such as H5N1, H5N6, and H7N9, have been reported to frequently infect humans, but acute encephalitis caused by HPAIV in humans has been rarely reported. We report the first critical case of acute encephalitis with mild pneumonia caused by the H5N6 virus. On January 25 of 2022, a 6-year-old girl with severe neurological symptoms was admitted to our hospital and rapidly developed into seizures and coma. Brain imaging showed abnormalities. Electroencephalogram (EEG) presented abnormal slow waves. Cerebrospinal fluid (CSF) contained elevated protein (1.64 g/L) and white cells (546 × 106/L). Laboratory investigations revealed abnormally elevated transaminases, lactate dehydrogenase, and cytokines in serum. A novel reassortant H5N6 virus was identified from the patient’s serum, CSF, and tracheal aspirate specimens. Phylogenic analysis indicated that this virus was a novel reassortant avian-origin influenza A (H5N6) virus that belonged to clade 2.3.4.4b. This patient was diagnosed with acute encephalitis and discharged from the hospital accompanied by a language barrier. An epidemiological investigation confirmed that wild waterfowls were the direct source of infection in this case. Our study highlights the urgent need to pay attention to acute encephalitis caused by HPAIV.
Collapse
Affiliation(s)
- Libing Zhang
- Department of Pediatrics of the Affiliated Hospital of Yangzhou University
| | - Kaituo Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University
| | - Qin Su
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College of Yangzhou University
| | - Xiao Chen
- Department of Pediatrics of the Affiliated Hospital of Yangzhou University
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University
| | - Qingfeng Li
- Department of Pediatrics of the Affiliated Hospital of Yangzhou University
| | - Wenlei Wang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College of Yangzhou University
| | - Xuhua Mao
- Department of Clinical Laboratory, the Affiliated Yixing Clinical School of Medical School of Yangzhou University
| | - Jinmei Xu
- Department of Pediatrics of the Affiliated Hospital of Yangzhou University
| | - Xin Zhou
- Yangzhou Center for Disease Control and Prevention; g Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Qin Xu
- Yangzhou Center for Disease Control and Prevention; g Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Le Zhou
- Yangzhou Center for Disease Control and Prevention; g Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University
| | - Pinghu Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College of Yangzhou University
| |
Collapse
|
17
|
Ye Y, Fan H, Li Q, Zhang Z, Miao P, Zhu J, Liu J, Zhang J, Liao M. Differential proteome response to H5N1 highly pathogenic avian influenza (HPAI) viruses infection in duck. Front Immunol 2022; 13:965454. [PMID: 36059479 PMCID: PMC9438030 DOI: 10.3389/fimmu.2022.965454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Ducks and wild aquatic birds are the natural reservoirs of avian influenza viruses. However, the host proteome response that causes disease in vivo by the H5N1 HPAI virus is still unclear. This study presented a comprehensive analysis of the proteome response in Muscovy duck lung tissue during 3 days of infection with either a highly virulent DK383 or an avirulent DK212. An unbiased strategy- isobaric tags for relative and absolute quantitation (iTRAQ) in conjunction with high-performance liquid chromatography with tandem mass spectrometry (HPLC-MS/MS) was utilized to investigate the infection mechanism. Pathways derived from analysis of 292 significantly altered proteins may contribute to the high pathogenic nature and disease progression of H5N1 viruses. Global proteome profiles indicated improved correlation with the virus titers and gene expression patterns between the two strains of the H5N1 virus. DK383 replicated more efficiently and induced a stronger response specific to severe disease. While proteins involved in the immune response of neutrophils were increased markedly by DK383, DK212 evoked a distinct response characterized by an increase in proteins involved in the maturation of dendritic cells, adhesion of phagocytes, and immune response of macrophages. The differentially activated Akt/mTOR/p70S6K pathway might involve in the host response to H5N1 viruses. Therefore, systematically integrated with datasets from primary genomic and virus titer results, proteomic analyses may help reveal the potential pathogenesis.
Collapse
Affiliation(s)
- Yu Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- College of Animal Technology and Science, Jiangxi Agricultural University, Nanchang, China
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China
| | - Qi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonoses Control and Prevention of Guangdong, Guangzhou, China
| | - Zhen Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Control and Prevention of Guangdong Higher Education Institutes, Guangzhou, China
| | - Peisi Miao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jun Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
| | - Jie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonoses Control and Prevention of Guangdong, Guangzhou, China
- Key Laboratory of Control and Prevention of Guangdong Higher Education Institutes, Guangzhou, China
| | - Jie Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China
- Key Laboratory of Zoonoses Control and Prevention of Guangdong, Guangzhou, China
- Key Laboratory of Control and Prevention of Guangdong Higher Education Institutes, Guangzhou, China
- *Correspondence: Ming Liao,
| |
Collapse
|
18
|
Genetic Determinants for Virulence and Transmission of the Panzootic Avian Influenza Virus H5N8 Clade 2.3.4.4 in Pekin Ducks. J Virol 2022; 96:e0014922. [PMID: 35670594 DOI: 10.1128/jvi.00149-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Waterfowl is the natural reservoir for avian influenza viruses (AIV), where the infection is mostly asymptomatic. In 2016, the panzootic high pathogenicity (HP) AIV H5N8 of clade 2.3.4.4B (designated H5N8-B) caused significant mortality in wild and domestic ducks, in stark contrast to the predecessor 2.3.4.4A virus from 2014 (designated H5N8-A). Here, we studied the genetic determinants for virulence and transmission of H5N8 clade 2.3.4.4 in Pekin ducks. While ducks inoculated with recombinant H5N8-A did not develop any clinical signs, H5N8-B-inoculated and cohoused ducks died after showing neurological signs. Swapping of the HA gene segments did not increase virulence of H5N8-A but abolished virulence and reduced systemic replication of H5N8-B. Only H5N8-A carrying H5N8-B HA, NP, and NS with or without NA exhibited high virulence in inoculated and contact ducks, similar to H5N8-B. Compared to H5N8-A, HA, NA, NS, and NP proteins of H5N8-B possess peculiar differences, which conferred increased receptor binding affinity, neuraminidase activity, efficiency to inhibit interferon-alpha induction, and replication in vitro, respectively. Taken together, this comprehensive study showed that HA is not the only virulence determinant of the panzootic H5N8-B in Pekin ducks, but NP, NS, and to a lesser extent NA were also necessary for the exhibition of high virulence in vivo. These proteins acted synergistically to increase receptor binding affinity, sialidase activity, interferon antagonism, and replication. This is the first ad-hoc study to investigate the mechanism underlying the high virulence of HPAIV in Pekin ducks. IMPORTANCE Since 2014, several waves of avian influenza virus (AIV) H5N8 of clade 2.3.4.4 occurred globally on unprecedented levels. Unlike viruses in the first wave in 2014-2015 (H5N8-A), viruses in 2015-2016 (H5N8-B) exhibited unusually high pathogenicity (HP) in wild and domestic ducks. Here, we found that the high virulence of H5N8-B in Pekin ducks could be attributed to multiple factors in combination, namely, hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), and nonstructural protein 1 (NS1). Compared to H5N8-A, H5N8-B possesses distinct genetic and biological properties including increased HA receptor-binding affinity and neuraminidase activity. Likewise, H5N8-B NS1 and NP were more efficient to inhibit interferon induction and enhance replication in primary duck cells, respectively. These results indicate the polygenic trait of virulence of HPAIV in domestic ducks and the altered biological properties of the HPAIV H5N8 clade 2.3.4.4B. These findings may explain the unusual high mortality in Pekin ducks during the panzootic H5N8 outbreaks.
Collapse
|
19
|
Durairaj K, Trinh TTT, Yun SY, Yeo SJ, Sung HW, 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:v14051001. [PMID: 35632743 PMCID: PMC9143286 DOI: 10.3390/v14051001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022] Open
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.
Collapse
Affiliation(s)
- Kaliannan Durairaj
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (K.D.); (S.-Y.Y.)
| | - Thuy-Tien Thi Trinh
- Institute of Endemic Diseases, Medical Research Center, Department of Tropical Medicine and Parasitology, Seoul National University, Seoul 03080, Korea;
| | - Su-Yeon Yun
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (K.D.); (S.-Y.Y.)
| | - Seon-Ju Yeo
- Department of Tropical Medicine and Parasitology, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
- Correspondence: (S.-J.Y.); (H.-W.S.); (H.P.)
| | - Haan-Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (S.-J.Y.); (H.-W.S.); (H.P.)
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (K.D.); (S.-Y.Y.)
- Correspondence: (S.-J.Y.); (H.-W.S.); (H.P.)
| |
Collapse
|
20
|
Zhao C, Guo J, Zeng X, Shi J, Deng G, Zhang Y, Wang Y, Ma Q, Gao X, Cui P, Liu L, Li X, Chen H. Novel H7N7 avian influenza viruses detected in migratory wild birds in eastern China between 2018 and 2020. Microbes Infect 2022; 24:105013. [DOI: 10.1016/j.micinf.2022.105013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022]
|
21
|
El-Shesheny R, Turner JCM, Walker D, Franks J, Seiler P, Barman S, Feeroz MM, Hasan MK, Akhtar S, Mukherjee N, Kercher L, McKenzie P, Webster RG, Webby RJ. Detection of a Novel Reassortant H9N9 Avian Influenza Virus in Free-Range Ducks in Bangladesh. Viruses 2021; 13:v13122357. [PMID: 34960626 PMCID: PMC8704232 DOI: 10.3390/v13122357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 01/10/2023] Open
Abstract
Wild aquatic birds are the primary natural reservoir for influenza A viruses (IAVs). In this study, an A(H9N9) influenza A virus (A/duck/Bangladesh/44493/2020) was identified via routine surveillance in free-range domestic ducks in Bangladesh. Phylogenetic analysis of hemagglutinin showed that the H9N9 virus belonged to the Y439-like lineage. The HA gene had the highest nucleotide identity to A/Bean Goose (Anser fabalis)/South Korea/KNU 2019-16/2019 (H9N2). The other seven gene segments clustered within the Eurasian lineage.
Collapse
Affiliation(s)
- Rabeh El-Shesheny
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Jasmine C. M. Turner
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - David Walker
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - John Franks
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Patrick Seiler
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Subrata Barman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Mohammed M. Feeroz
- Department of Zoology, Jahangirnagar University, Savar 1342, Bangladesh; (M.M.F.); (M.K.H.); (S.A.)
| | - Md Kamrul Hasan
- Department of Zoology, Jahangirnagar University, Savar 1342, Bangladesh; (M.M.F.); (M.K.H.); (S.A.)
| | - Sharmin Akhtar
- Department of Zoology, Jahangirnagar University, Savar 1342, Bangladesh; (M.M.F.); (M.K.H.); (S.A.)
| | - Nabanita Mukherjee
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Lisa Kercher
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Pamela McKenzie
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Robert G. Webster
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.E.-S.); (J.C.M.T.); (D.W.); (J.F.); (P.S.); (S.B.); (N.M.); (L.K.); (P.M.); (R.G.W.)
- Correspondence:
| |
Collapse
|
22
|
Vijayakumar P, Raut AA, Chingtham S, Murugkar HV, Kulkarni DD, Sood R, Singh VP, Mishra A. Proteomic analysis of differential expression of lung proteins in response to highly pathogenic avian influenza virus infection in chickens. Arch Virol 2021; 167:141-152. [PMID: 34786609 DOI: 10.1007/s00705-021-05287-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/16/2021] [Indexed: 12/21/2022]
Abstract
Elucidation of the molecular pathogenesis underlying virus-host interactions is important for the development of new diagnostic and therapeutic strategies against highly pathogenic avian influenza (HPAI) virus infection in chickens. However, the pathogenesis of HPAI virus in chickens is not completely understood. To identify the intracellular signaling pathways and critical host proteins associated with influenza pathogenesis, we analyzed the lung proteome of a chicken infected with HPAI H5N1 virus (A/duck/India/02CA10/2011/Agartala). Mass spectrometry data sets were searched against the chicken UniProt reference database. At the local false discovery rate level of 5%, a total of 3313 proteins with the presence of at least one unique peptide were identified in the chicken lung proteome datasets. Differential expression analysis of these proteins showed that 247 and 1754 proteins were downregulated at 12 h and 48 h postinfection, respectively. We observed expression of proteins of the predominant signaling pathways, including Toll-like receptors (TLRs), retinoic acid-inducible gene I-like receptors (RLRs), NOD-like receptors (NLRs), and JAK-STAT signaling. Activation of these pathways is associated with the cytokine storm effect and thus may be the cause of the severity of HPAI H5N1 infection in chickens. We also observed the expression of myeloid differentiation primary response protein (MyD88), inhibitor of nuclear factor kappa B kinase subunit beta (IKBKB), interleukin 1 receptor associated kinase 4 (IRAK4), RELA proto-oncogene NF-κB subunit (RELA), and mitochondrial antiviral signaling protein (MAVS), which are involved in critical signaling pathways, as well as other, less-commonly identified proteins such as hepatocyte nuclear factor 4 alpha (HNF4A), ELAV-like RNA binding protein 1 (ELAVL1), fibronectin 1 (FN1), COP9 signalosome subunit 5 (COPS5), cullin 1 (CUL1), breast cancer type 1 susceptibility protein (BRCA1), and the FYN proto-oncogene Src family tyrosine kinase (FYN) as main hub proteins that might play important roles in influenza pathogenesis in chickens. In summary, we identified the signaling pathways and the proteomic determinants associated with disease pathogenesis in chickens infected with HPAI H5N1 virus.
Collapse
Affiliation(s)
- Periyasamy Vijayakumar
- Pathogenomics Laboratory, ICAR-National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India.,Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Orathanadu, 614625, Tamil Nadu, India
| | - Ashwin Ashok Raut
- Pathogenomics Laboratory, ICAR-National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India
| | - Santhalembi Chingtham
- Pathogenomics Laboratory, ICAR-National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India
| | - Harshad V Murugkar
- ICAR -National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India
| | - Diwakar D Kulkarni
- ICAR -National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India
| | - Richa Sood
- ICAR -National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India
| | - Vijendra Pal Singh
- ICAR -National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India
| | - Anamika Mishra
- Pathogenomics Laboratory, ICAR-National Institute of High-Security Animal Diseases, OIE Reference lab for Avian Influenza, Bhopal, 462021, Madhya Pradesh, India.
| |
Collapse
|
23
|
Duong BT, Bal J, Sung HW, Yeo SJ, 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:v13112260. [PMID: 34835066 PMCID: PMC8623559 DOI: 10.3390/v13112260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022] Open
Abstract
Avian influenza virus (AIV) subtypes H5 and H7, possessing the ability to mutate spontaneously from low pathogenic (LP) to highly pathogenic (HP) variants, are major concerns for enormous socio-economic losses in the poultry industry, as well as for fatal human infections. Through antigenic drift and shift, genetic reassortments of the genotypes pose serious threats of increased virulence and pathogenicity leading to potential pandemics. In this study, we isolated the H7-subtype AIVs circulating in the Republic of Korea during 2018–2019, and perform detailed molecular analysis to study their circulation, evolution, and possible emergence as a zoonotic threat. Phylogenetic and nucleotide sequence analyses of these isolates revealed their distribution into two distinct clusters, with the HA gene sharing the highest nucleotide identity with either the A/common teal/Shanghai/CM1216/2017, isolated from wild birds in Shanghai, China, or the A/duck/Shimane/2014, isolated from Japan. Mutations were found in HA (S138A (H3 numbering)), M1 (N30D and T215A), NS1 (P42S), PB2 (L89V), and PA (H266R and F277S) proteins—the mutations had previously been reported to be related to mammalian adaptation and changes in the virulence of AIVs. Taken together, the results firmly put forth the demand for routine surveillance of AIVs in wild birds to prevent possible pandemics arising from reassortant AIVs.
Collapse
Affiliation(s)
- Bao Tuan Duong
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (B.T.D.); (J.B.)
| | - Jyotiranjan Bal
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (B.T.D.); (J.B.)
| | - Haan Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon-si 24341, Korea
- Correspondence: (H.W.S.); (S.-J.Y.); (H.P.)
| | - Seon-Ju Yeo
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: (H.W.S.); (S.-J.Y.); (H.P.)
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (B.T.D.); (J.B.)
- Correspondence: (H.W.S.); (S.-J.Y.); (H.P.)
| |
Collapse
|
24
|
Nguyen ATV, Hoang VT, Sung HW, Yeo SJ, Park H. Genetic Characterization and Pathogenesis of Three Novel Reassortant H5N2 Viruses in South Korea, 2018. Viruses 2021; 13:v13112192. [PMID: 34834997 PMCID: PMC8619638 DOI: 10.3390/v13112192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 12/03/2022] Open
Abstract
The outbreaks of H5N2 avian influenza viruses have occasionally caused the death of thousands of birds in poultry farms. Surveillance during the 2018 winter season in South Korea revealed three H5N2 isolates in feces samples collected from wild birds (KNU18-28: A/Wild duck/South Korea/KNU18-28/2018, KNU18-86: A/Bean Goose/South Korea/KNU18-86/2018, and KNU18-93: A/Wild duck/South Korea/KNU18-93/2018). Phylogenetic tree analysis revealed that these viruses arose from reassortment events among various virus subtypes circulating in South Korea and other countries in the East Asia–Australasian Flyway. The NS gene of the KNU18-28 and KNU18-86 isolates was closely related to that of China’s H10N3 strain, whereas the KNU18-93 strain originated from the H12N2 strain in Japan, showing two different reassortment events and different from a low pathogenic H5N3 (KNU18-91) virus which was isolated at the same day and same place with KNU18-86 and KNU18-93. These H5N2 isolates were characterized as low pathogenic avian influenza viruses. However, many amino acid changes in eight gene segments were identified to enhance polymerase activity and increase adaptation and virulence in mice and mammals. Experiments reveal that viral replication in MDCK cells was quite high after 12 hpi, showing the ability to replicate in mouse lungs. The hematoxylin and eosin-stained (H&E) lung sections indicated different degrees of pathogenicity of the three H5N2 isolates in mice compared with that of the control H1N1 strain. The continuing circulation of these H5N2 viruses may represent a potential threat to mammals and humans. Our findings highlight the need for intensive surveillance of avian influenza virus circulation in South Korea to prevent the risks posed by these reassortment viruses to animal and public health.
Collapse
Affiliation(s)
- Anh Thi Viet Nguyen
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (A.T.V.N.); (V.T.H.)
| | - Vui Thi Hoang
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (A.T.V.N.); (V.T.H.)
| | - Haan Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Seon-Ju Yeo
- Department of Tropical Medicine and Parasitology, College of Medicine, Seoul National University, Seoul 03080, Korea
- Correspondence: (S.-J.Y.); (H.P.)
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (A.T.V.N.); (V.T.H.)
- Correspondence: (S.-J.Y.); (H.P.)
| |
Collapse
|
25
|
Wan Z, Kan Q, Zhao Z, Shao H, Deliberto TJ, Wan XF, Qin A, Ye J. Characterization of Subtype H6 Avian Influenza A Viruses Isolated From Wild Birds in Poyang Lake, China. Front Vet Sci 2021; 8:685399. [PMID: 34589532 PMCID: PMC8473872 DOI: 10.3389/fvets.2021.685399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Subtype H6 avian influenza A viruses (IAVs) are enzootic and genetically diverse in both domestic poultry and wild waterfowl and may cause spillovers in both pigs and humans. Thus, it is important to understand the genetic diversity of H6 IAVs in birds and their zoonotic potential. Compared with that in domestic poultry, the genetic diversity of H6 viruses in wild birds in China has not been well-understood. In this study, five H6 viruses were isolated from wild birds in Poyang Lake, China, and genetic analyses showed that these isolates are clustered into four genotypes associated with reassortments among avian IAVs from domestic poultry and wild birds in China and those from Eurasia and North America and that these viruses exhibited distinct phenotypes in growth kinetics analyses with avian and mammalian cells lines and in mouse challenge experiments. Of interest is that two H6 isolates from the Eurasian teal replicated effectively in the mouse lung without prior adaptation, whereas the other three did not. Our study suggested that there are variations in the mammalian viral replication efficiency phenotypic among genetically diverse H6 IAVs in wild birds and that both intra- and inter-continental movements of IAVs through wild bird migration may facilitate the emergence of novel H6 IAV reassortants with the potential for replicating in mammals, including humans. Continued surveillance to monitor the diversity of H6 IAVs in wild birds is necessary to increase our understanding of the natural history of IAVs.
Collapse
Affiliation(s)
- Zhimin Wan
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Key Laboratory for Avian Preventive Medicine, Ministry of Education, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Qiuqi Kan
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Key Laboratory for Avian Preventive Medicine, Ministry of Education, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Zhehong Zhao
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Key Laboratory for Avian Preventive Medicine, Ministry of Education, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Hongxia Shao
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Key Laboratory for Avian Preventive Medicine, Ministry of Education, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Thomas J Deliberto
- National Wildlife Disease Program, Wildlife Services, Animal and Plant Health Inspection Service, U.S. Department of Agriculture, Fort Collins, CO, United States
| | - Xiu-Feng Wan
- University of Missouri Center for Influenza and Emerging infectious Diseases, University of Missouri, Columbia, MO, United States.,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States.,Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.,Department of Electrical Engineering and Computer Science, College of Engineering, University of Missouri, Columbia, MO, United States
| | - Aijian Qin
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Key Laboratory for Avian Preventive Medicine, Ministry of Education, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jianqiang Ye
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Key Laboratory for Avian Preventive Medicine, Ministry of Education, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
26
|
Multiple Gene Segments Are Associated with Enhanced Virulence of Clade 2.3.4.4 H5N8 Highly Pathogenic Avian Influenza Virus in Mallards. J Virol 2021; 95:e0095521. [PMID: 34232725 DOI: 10.1128/jvi.00955-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Highly pathogenic avian influenza (HPAI) viruses from the H5Nx Goose/Guangdong/96 lineage continue to cause outbreaks in domestic and wild bird populations. Two distinct genetic groups of H5N8 HPAI viruses, hemagglutinin (HA) clades 2.3.4.4A and 2.3.4.4B, caused intercontinental outbreaks in 2014 to 2015 and 2016 to 2017, respectively. Experimental infections using viruses from these outbreaks demonstrated a marked difference in virulence in mallards, with the H5N8 virus from 2014 causing mild clinical disease and the 2016 H5N8 virus causing high mortality. To assess which gene segments are associated with enhanced virulence of H5N8 HPAI viruses in mallards, we generated reassortant viruses with 2014 and 2016 viruses. For single-segment reassortants in the genetic backbone of the 2016 virus, pathogenesis experiments in mallards revealed that morbidity and mortality were reduced for all eight single-segment reassortants compared to the parental 2016 virus, with significant reductions in mortality observed with the polymerase basic protein 2 (PB2), nucleoprotein (NP), and matrix (M) reassortants. No differences in morbidity and mortality were observed with reassortants that either have the polymerase complex segments or the HA and neuraminidase (NA) segments of the 2016 virus in the genetic backbone of the 2014 virus. In vitro assays showed that the NP and polymerase acidic (PA) segments of the 2014 virus lowered polymerase activity when combined with the polymerase complex segments of the 2016 virus. Furthermore, the M segment of the 2016 H5N8 virus was linked to filamentous virion morphology. Phylogenetic analyses demonstrated that gene segments related to the more virulent 2016 H5N8 virus have persisted in the contemporary H5Nx HPAI gene pool until 2020. IMPORTANCE Outbreaks of H5Nx HPAI viruses from the goose/Guangdong/96 lineage continue to occur in many countries and have resulted in substantial impact on wild birds and poultry. Epidemiological evidence has shown that wild waterfowl play a major role in the spread of these viruses. While HPAI virus infection in gallinaceous species causes high mortality, a wide range of disease outcomes has been observed in waterfowl species. In this study, we examined which gene segments contribute to severe disease in mallards infected with H5N8 HPAI viruses. No virus gene was solely responsible for attenuating the high virulence of a 2016 H5N8 virus, but the PB2, NP, and M segments significantly reduced mortality. The findings herein advance our knowledge on the pathobiology of avian influenza viruses in waterfowl and have potential implications on the ecology and epidemiology of H5Nx HPAI in wild bird populations.
Collapse
|
27
|
Preferential Selection and Contribution of Non-Structural Protein 1 (NS1) to the Efficient Transmission of Panzootic Avian Influenza H5N8 Virus Clades 2.3.4.4A and B in Chickens and Ducks. J Virol 2021; 95:e0044521. [PMID: 34160261 DOI: 10.1128/jvi.00445-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Highly pathogenic avian influenza virus H5N8 clade 2.3.4.4 caused outbreaks in poultry at an unprecedented global scale. The virus was spread by wild birds in Asia in two waves: clade 2.3.4.4A in 2014/2015 and clade 2.3.4.4B from 2016 up to today. Both clades were highly virulent in chickens, but only clade B viruses exhibited high virulence in ducks. Viral factors which contribute to virulence and transmission of these panzootic H5N8 2.3.4.4 viruses are largely unknown. The NS1 protein, typically composed of 230 amino acids (aa), is a multifunctional protein which is also a pathogenicity factor. Here, we studied the evolutionary trajectory of H5N8 NS1 proteins from 2013 to 2019 and their role in the fitness of H5N8 viruses in chickens and ducks. Sequence analysis and in vitro experiments indicated that clade 2.3.4.4A and clade 2.3.4.4B viruses have a preference for NS1 of 237 aa and 217 aa, respectively, over NS1 of 230 aa. NS217 was exclusively seen in domestic and wild birds in Europe. The extension of the NS1 C terminus (CTE) of clade B virus reduced virus transmission and replication in chickens and ducks and partially impaired the systemic tropism to the endothelium in ducks. Conversely, lower impact on fitness of clade A virus was observed. Remarkably, the NS1 of clade A and clade B, regardless of length, was efficient in blocking interferon (IFN) induction in infected chickens, and changes in the NS1 C terminus reduced the efficiency for interferon antagonism. Together, the NS1 C terminus contributes to the efficient transmission and high fitness of H5N8 viruses in chickens and ducks. IMPORTANCE The panzootic H5N8 highly pathogenic avian influenza viruses of clade 2.3.4.4A and 2.3.4.4B devastated the poultry industry globally. Clade 2.3.4.4A was predominant in 2014/2015 while clade 2.3.4.4B was widely spread in 2016/2017. The two clades exhibited different pathotypes in ducks. Virus factors contributing to virulence and transmission are largely unknown. The NS1 protein is typically composed of 230 amino acids (aa) and is an essential interferon (IFN) antagonist. Here, we found that the NS1 protein of clade 2.3.4.4A preferentially evolved toward long NS1 with 237 aa, while clade 2.3.4.4B evolved toward shorter NS1 with 217 aa (exclusively found in Europe) due to stop codons in the C terminus (CTE). We showed that the NS1 CTE of H5N8 is required for efficient virus replication, transmission, and endotheliotropism in ducks. In chickens, H5N8 NS1 evolved toward higher efficiency to block IFN response. These findings may explain the preferential pattern for short NS1 and high fitness of the panzootic H5N8 in birds.
Collapse
|
28
|
Identification and molecular characterization of H9N2 viruses carrying multiple mammalian adaptation markers in resident birds in central-western wetlands in India. INFECTION GENETICS AND EVOLUTION 2021; 94:105005. [PMID: 34293481 DOI: 10.1016/j.meegid.2021.105005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/03/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022]
Abstract
We report here a targeted risk-based study to investigate the presence of influenza A viruses at the migratory-wild-domestic bird interface across the major wetlands of central India's Maharashtra state during the winter migration season. The H9N2 viruses have been isolated and confirmed in 3.86% (33/854) of the fecal samples of resident birds. To investigate the genetic pools of H9N2 circulating in resident birds, we sequenced two isolates of H9N2 from distant wetlands. Sequence and phylogenetic analyses have shown that these viruses are triple reassortants, with HA, NA, NP, and M genes belonging to G1 sub-lineage (A/quail/Hong Kong/G1/1997), PB2, PB1, and NS genes originating from the prototype Eurasian lineage (A/mallard/France/090360/2009) and PA gene deriving from Y439/Korean-like (A/duck/Hong Kong/Y439/97) sub-lineage. It was confirmed not only that four of their gene segments had a high genetic association with the zoonotic H9N2 virus, A/Human/India/TCM2581/2019, but also that they had many molecular markers associated with mammalian adaptation and enhanced virulence in mammals including the unique multiple basic amino acids, KSKR↓GLF at the HA cleavage site, and analog N-and O-glycosylation patterns on HA with that of the zoonotic H9N2 virus. Furthermore, future experiments would be to characterize these isolates biologically to address the public health concern. Importantly, due to the identification of these viruses at a strategic geographical location in India (a major stop-over point in the Central Asian flyway), these novel viruses also pose a possible threat to be exported to other regions via migratory/resident birds. Consequently, systematic investigation and active monitoring are a prerequisite for identifying and preventing the spread of viruses of zoonotic potential by enforcing strict biosecurity measures.
Collapse
|
29
|
Waters K, Wan HJ, Han L, Xue J, Ykema M, Tao YJ, Wan XF. Variations outside the conserved motifs of PB1 catalytic active site may affect replication efficiency of the RNP complex of influenza A virus. Virology 2021; 559:145-155. [PMID: 33887645 PMCID: PMC8579824 DOI: 10.1016/j.virol.2021.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/31/2021] [Accepted: 04/05/2021] [Indexed: 12/24/2022]
Abstract
PB1 functions as the catalytic subunit of influenza virus RNA polymerase complex and plays an essential role in viral RNA transcription and replication. To determine plasticity in the PB1 enzymatic site and map catalytically important residues, 658 mutants were constructed, each with one to seven mutations in the enzymatic site of PB1. The polymerase activities of these mutants were quantified using a minigenome assay, and polymerase activity-associated residues were identified using sparse learning. Results showed that polymerase activities are affected by the residues not only within the conserved motifs, but also across the inter-motif regions of PB1, and the latter are primarily located at the base of the palm domain, a region that is conserved in avian PB1 but with high sequence diversity in swine PB1. Our results suggest that mutations outside the PB1 conserved motifs may affect RNA replication and could be associated with influenza virus host adaptation.
Collapse
Affiliation(s)
- Kaitlyn Waters
- Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA; Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Hamilton J Wan
- Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA; Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Lei Han
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Jianli Xue
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Matthew Ykema
- Department of BioSciences, Rice University, Houston, TX, 77251, USA
| | - Yizhi J Tao
- Department of BioSciences, Rice University, Houston, TX, 77251, USA
| | - Xiu-Feng Wan
- Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA; Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA; Department of Electrical Engineering & Computer Science, College of Engineering, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
30
|
Health monitoring in birds using bio-loggers and whole blood transcriptomics. Sci Rep 2021; 11:10815. [PMID: 34031452 PMCID: PMC8144624 DOI: 10.1038/s41598-021-90212-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Monitoring and early detection of emerging infectious diseases in wild animals is of crucial global importance, yet reliable ways to measure immune status and responses are lacking for animals in the wild. Here we assess the usefulness of bio-loggers for detecting disease outbreaks in free-living birds and confirm detailed responses using leukocyte composition and large-scale transcriptomics. We simulated natural infections by viral and bacterial pathogens in captive mallards (Anas platyrhynchos), an important natural vector for avian influenza virus. We show that body temperature, heart rate and leukocyte composition change reliably during an acute phase immune response. Using genome-wide gene expression profiling of whole blood across time points we confirm that immunostimulants activate pathogen-specific gene regulatory networks. By reporting immune response related changes in physiological and behavioural traits that can be studied in free-ranging populations, we provide baseline information with importance to the global monitoring of zoonotic diseases.
Collapse
|
31
|
Sun N, Li C, Li XF, Deng YQ, Jiang T, Zhang NN, Zu S, Zhang RR, Li L, Chen X, Liu P, Gold S, Lu N, Du P, Wang J, Qin CF, Cheng G. Type-IInterferon-Inducible SERTAD3 Inhibits Influenza A Virus Replication by Blocking the Assembly of Viral RNA Polymerase Complex. Cell Rep 2021; 33:108342. [PMID: 33147462 DOI: 10.1016/j.celrep.2020.108342] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 01/08/2023] Open
Abstract
Influenza A virus (IAV) infection stimulates a type I interferon (IFN-I) response in host cells that exerts antiviral effects by inducing the expression of hundreds of IFN-stimulated genes (ISGs). However, most ISGs are poorly studied for their roles in the infection of IAV. Herein, we demonstrate that SERTA domain containing 3 (SERTAD3) has a significant inhibitory effect on IAV replication in vitro. More importantly, Sertad3-/- mice develop more severe symptoms upon IAV infection. Mechanistically, we find SERTAD3 reduces IAV replication through interacting with viral polymerase basic protein 2 (PB2), polymerase basic protein 1 (PB1), and polymerase acidic protein (PA) to disrupt the formation of the RNA-dependent RNA polymerase (RdRp) complex. We further identify an 8-amino-acid peptide of SERTAD3 as a minimum interacting motif that can disrupt RdRp complex formation and inhibit IAV replication. Thus, our studies not only identify SERTAD3 as an antiviral ISG, but also provide the mechanism of potential application of SERTAD3-derived peptide in suppressing influenza replication.
Collapse
Affiliation(s)
- Nina Sun
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China; Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of System Medicine, Suzhou, Jiangsu 215123, China
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Department of Pathology, Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yong-Qiang Deng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Tao Jiang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Na-Na Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Shulong Zu
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China; Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of System Medicine, Suzhou, Jiangsu 215123, China
| | - Rong-Rong Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Lili Li
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of System Medicine, Suzhou, Jiangsu 215123, China
| | - Xiang Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ping Liu
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Sarah Gold
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ning Lu
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
| | - Peishuang Du
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
| | - Jingfeng Wang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of System Medicine, Suzhou, Jiangsu 215123, China; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Genetic Characterization and Pathogenesis of Avian Influenza Virus H7N3 Isolated from Spot-Billed Ducks in South Korea, Early 2019. Viruses 2021; 13:v13050856. [PMID: 34067187 PMCID: PMC8151380 DOI: 10.3390/v13050856] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 11/29/2022] Open
Abstract
Low-pathogenicity avian influenza viruses (LPAIV) introduced by migratory birds circulate in wild birds and can be transmitted to poultry. These viruses can mutate to become highly pathogenic avian influenza viruses causing severe disease and death in poultry. In March 2019, an H7N3 avian influenza virus—A/Spot-billed duck/South Korea/WKU2019-1/2019 (H7N3)—was isolated from spot-billed ducks in South Korea. This study aimed to evaluate the phylogenetic and mutational analysis of this isolate. Molecular analysis revealed that the genes for HA (hemagglutinin) and NA (neuraminidase) of this strain belonged to the Central Asian lineage, whereas genes for other internal proteins such as polymerase basic protein 1 (PB1), PB2, nucleoprotein, polymerase acidic protein, matrix protein, and non-structural protein belonged to that of the Korean lineage. In addition, a monobasic amino acid (PQIEPR/GLF) at the HA cleavage site, and the non-deletion of the stalk region in the NA gene indicated that this isolate was a typical LPAIV. Nucleotide sequence similarity analysis of HA revealed that the highest homology (99.51%) of this isolate is to that of A/common teal/Shanghai/CM1216/2017 (H7N7), and amino acid sequence of NA (99.48%) was closely related to that of A/teal/Egypt/MB-D-487OP/2016 (H7N3). An in vitro propagation of the A/Spot-billed duck/South Korea/WKU2019-1/2019 (H7N3) virus showed highest (7.38 Log10 TCID50/mL) virus titer at 60 h post-infection, and in experimental mouse lungs, the virus was detected at six days’ post-infection. Our study characterizes genetic mutations, as well as pathogenesis in both in vitro and in vivo model of a new Korea H7N3 viruses in 2019, carrying multiple potential mutations to become highly pathogenic and develop an ability to infect humans; thus, emphasizing the need for routine surveillance of avian influenza viruses in wild birds.
Collapse
|
33
|
Mercan Y, Atim G, Kayed AE, Azbazdar ME, Kandeil A, Ali MA, Rubrum A, McKenzie P, Webby RJ, Erima B, Wabwire-Mangen F, Ukuli QA, Tugume T, Byarugaba DK, Kayali G, Ducatez MF, Koçer ZA. Molecular Characterization of Closely Related H6N2 Avian Influenza Viruses Isolated from Turkey, Egypt, and Uganda. Viruses 2021; 13:v13040607. [PMID: 33918166 PMCID: PMC8065897 DOI: 10.3390/v13040607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023] Open
Abstract
Genetic analysis of circulating avian influenza viruses (AIVs) in wild birds at different geographical regions during the same period could improve our knowledge about virus transmission dynamics in natural hosts, virus evolution as well as zoonotic potential. Here, we report the genetic and molecular characterization of H6N2 influenza viruses isolated from migratory birds in Turkey, Egypt, and Uganda during 2017–2018. The Egyptian and Turkish isolates were genetically closer to each other than they were to the virus isolated from Uganda. Our results also suggest that multiple reassortment events were involved in the genesis of the isolated viruses. All viruses contained molecular markers previously associated with increased replication and/or pathogenicity in mammals. The results of this study indicate that H6N2 viruses carried by migratory birds on the West Asian/East African and Mediterranean/Black Sea flyways have the potential to transmit to mammals including humans. Additionally, adaptation markers in these viruses indicate the potential risk for poultry, which also increases the possibility of human exposure to these viruses.
Collapse
Affiliation(s)
- Yavuz Mercan
- Emerging Viral Diseases Laboratory, Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; (Y.M.); (M.E.A.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Izmir, Turkey
| | - Gladys Atim
- Makerere University Walter Reed Project, P.O. Box 7062 Kampala, Uganda; (G.A.); (B.E.); (F.W.-M.); (Q.A.U.); (T.T.); (D.K.B.)
| | - Ahmed E. Kayed
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12311, Egypt; (A.E.K.); (A.K.); (M.A.A.)
| | - M. Ekin Azbazdar
- Emerging Viral Diseases Laboratory, Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; (Y.M.); (M.E.A.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Izmir, Turkey
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12311, Egypt; (A.E.K.); (A.K.); (M.A.A.)
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12311, Egypt; (A.E.K.); (A.K.); (M.A.A.)
| | - Adam Rubrum
- St Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (P.M.); (R.J.W.)
| | - Pamela McKenzie
- St Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (P.M.); (R.J.W.)
| | - Richard J. Webby
- St Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (P.M.); (R.J.W.)
| | - Bernard Erima
- Makerere University Walter Reed Project, P.O. Box 7062 Kampala, Uganda; (G.A.); (B.E.); (F.W.-M.); (Q.A.U.); (T.T.); (D.K.B.)
| | - Fred Wabwire-Mangen
- Makerere University Walter Reed Project, P.O. Box 7062 Kampala, Uganda; (G.A.); (B.E.); (F.W.-M.); (Q.A.U.); (T.T.); (D.K.B.)
- School of Public Health, Makerere University, P.O. Box 7062 Kampala, Uganda
| | - Qouilazoni A. Ukuli
- Makerere University Walter Reed Project, P.O. Box 7062 Kampala, Uganda; (G.A.); (B.E.); (F.W.-M.); (Q.A.U.); (T.T.); (D.K.B.)
| | - Titus Tugume
- Makerere University Walter Reed Project, P.O. Box 7062 Kampala, Uganda; (G.A.); (B.E.); (F.W.-M.); (Q.A.U.); (T.T.); (D.K.B.)
| | - Denis K. Byarugaba
- Makerere University Walter Reed Project, P.O. Box 7062 Kampala, Uganda; (G.A.); (B.E.); (F.W.-M.); (Q.A.U.); (T.T.); (D.K.B.)
- College of Veterinary Medicine, Makerere University, P.O. Box 7062 Kampala, Uganda
| | - Ghazi Kayali
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas, Houston, TX 77030, USA;
- Human Link, Dubai, United Arab Emirates
| | | | - Zeynep A. Koçer
- Emerging Viral Diseases Laboratory, Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; (Y.M.); (M.E.A.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Izmir, Turkey
- Correspondence: ; Tel.: +90-232-299-4165
| |
Collapse
|
34
|
Youk SS, Leyson CM, Seibert BA, Jadhao S, Perez DR, Suarez DL, Pantin-Jackwood MJ. Mutations in PB1, NP, HA, and NA Contribute to Increased Virus Fitness of H5N2 Highly Pathogenic Avian Influenza Virus Clade 2.3.4.4 in Chickens. J Virol 2021; 95:JVI.01675-20. [PMID: 33268526 PMCID: PMC8092828 DOI: 10.1128/jvi.01675-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 11/19/2020] [Indexed: 12/26/2022] Open
Abstract
The H5N8 highly pathogenic avian influenza (HPAI) clade 2.3.4.4 virus spread to North America by wild birds and reassorted to generate the H5N2 HPAI virus that caused the poultry outbreak in the United States in 2015. In previous studies, we showed that H5N2 viruses isolated from poultry in the later stages of the outbreak had higher infectivity and transmissibility in chickens than the wild bird index H5N2 virus. Here, we determined the genetic changes that contributed to the difference in host virus fitness by analyzing sequence data from all of the viruses detected during the H5N2 outbreak, and studying the pathogenicity of reassortant viruses generated with the index wild bird virus and a chicken virus from later in the outbreak. Viruses with the wild bird virus backbone and either PB1, NP, or the entire polymerase complex of the chicken isolate, caused higher and earlier mortality in chickens, with three mutations (PB1 E180D, M317V, and NP I109T) identified to increase polymerase activity in chicken cells. The reassortant virus with the HA and NA from the chicken virus, where mutations in functionally known gene regions were acquired as the virus circulated in turkeys (HA S141P and NA S416G) and later in chickens (HA M66I, L322Q), showed faster virus growth, bigger plaque size and enhanced heat persistence in vitro, and increased pathogenicity and transmissibility in chickens. Collectively, these findings demonstrate an evolutionary pathway in which a HPAI virus from wild birds can accumulate genetic changes to increase fitness in poultry.IMPORTANCE H5Nx highly pathogenic avian influenza (HPAI) viruses of the A/goose/Guangdong/1/96 lineage continue to circulate widely affecting both poultry and wild birds. These viruses continue to change and reassort, which affects their fitness to different avian hosts. In this study, we defined mutations associated with increased virus fitness in chickens as the clade 2.3.4.4. H5N2 HPAI virus circulated in different avian species. We identified mutations in the PB1, NP, HA, and NA virus proteins that were highly conserved in the poultry isolates and contributed to the adaptation of this virus in chickens. This knowledge is important for understanding the epidemiology of H5Nx HPAI viruses and specifically the changes related to adaptation of these viruses in poultry.
Collapse
Affiliation(s)
- Sung-Su Youk
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Christina M Leyson
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Brittany A Seibert
- Department of Population Health, University of Georgia, Poultry Diagnostic and Research Center, Athens, Georgia, USA
| | - Samadhan Jadhao
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
- Department of Pediatrics, School of Medicine, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Daniel R Perez
- Department of Population Health, University of Georgia, Poultry Diagnostic and Research Center, Athens, Georgia, USA
| | - David L Suarez
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Mary J Pantin-Jackwood
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| |
Collapse
|
35
|
Genetic Characteristics of Avian Influenza Virus Isolated from Wild Birds in South Korea, 2019-2020. Viruses 2021; 13:v13030381. [PMID: 33673635 PMCID: PMC7997295 DOI: 10.3390/v13030381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 11/17/2022] Open
Abstract
Wild aquatic birds, a natural reservoir of avian influenza viruses (AIVs), transmit AIVs to poultry farms, causing huge economic losses. Therefore, the prevalence and genetic characteristics of AIVs isolated from wild birds in South Korea from October 2019 to March 2020 were investigated and analyzed. Fresh avian fecal samples (3256) were collected by active monitoring of 11 wild bird habitats. Twenty-eight AIVs were isolated. Seven HA and eight NA subtypes were identified. All AIV hosts were Anseriformes species. The HA cleavage site of 20 representative AIVs was encoded by non-multi-basic amino acid sequences. Phylogenetic analysis of the eight segment genes of the AIVs showed that most genes clustered within the Eurasian lineage. However, the HA gene of H10 viruses and NS gene of four viruses clustered within the American lineage, indicating intercontinental reassortment of AIVs. Representative viruses likely to infect mammals were selected and evaluated for pathogenicity in mice. JB21-58 (H5N3), JB42-93 (H9N2), and JB32-81 (H11N2) were isolated from the lungs, but JB31-69 (H11N9) was not isolated from the lungs until the end of the experiment at 14 dpi. None of infected mice showed clinical sign and histopathological change in the lung. In addition, viral antigens were not detected in lungs of all mice at 14 dpi. These data suggest that LPAIVs derived from wild birds are unlikely to be transmitted to mammals. However, because LPAIVs can reportedly infect mammals, including humans, continuous surveillance and monitoring of AIVs are necessary, despite their low pathogenicity.
Collapse
|
36
|
Verhagen JH, Fouchier RAM, Lewis N. Highly Pathogenic Avian Influenza Viruses at the Wild-Domestic Bird Interface in Europe: Future Directions for Research and Surveillance. Viruses 2021; 13:212. [PMID: 33573231 PMCID: PMC7912471 DOI: 10.3390/v13020212] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) outbreaks in wild birds and poultry are no longer a rare phenomenon in Europe. In the past 15 years, HPAI outbreaks-in particular those caused by H5 viruses derived from the A/Goose/Guangdong/1/1996 lineage that emerged in southeast Asia in 1996-have been occuring with increasing frequency in Europe. Between 2005 and 2020, at least ten HPAI H5 incursions were identified in Europe resulting in mass mortalities among poultry and wild birds. Until 2009, the HPAI H5 virus outbreaks in Europe were caused by HPAI H5N1 clade 2.2 viruses, while from 2014 onwards HPAI H5 clade 2.3.4.4 viruses dominated outbreaks, with abundant genetic reassortments yielding subtypes H5N1, H5N2, H5N3, H5N4, H5N5, H5N6 and H5N8. The majority of HPAI H5 virus detections in wild and domestic birds within Europe coincide with southwest/westward fall migration and large local waterbird aggregations during wintering. In this review we provide an overview of HPAI H5 virus epidemiology, ecology and evolution at the interface between poultry and wild birds based on 15 years of avian influenza virus surveillance in Europe, and assess future directions for HPAI virus research and surveillance, including the integration of whole genome sequencing, host identification and avian ecology into risk-based surveillance and analyses.
Collapse
Affiliation(s)
- Josanne H. Verhagen
- Department of Viroscience, Erasmus Medical Center, 3015 GD Rotterdam, Zuid-Holland, The Netherlands; (J.H.V.); (R.A.M.F.)
| | - Ron A. M. Fouchier
- Department of Viroscience, Erasmus Medical Center, 3015 GD Rotterdam, Zuid-Holland, The Netherlands; (J.H.V.); (R.A.M.F.)
| | - Nicola Lewis
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield AL9 7TA, Hertfordshire, UK
| |
Collapse
|
37
|
Li M, Qi W, Chang Q, Chen R, Zhen D, Liao M, Wen J, Deng Y. Influenza A virus protein PA-X suppresses host Ankrd17-mediated immune responses. Microbiol Immunol 2021; 65:48-59. [PMID: 33241870 DOI: 10.1111/1348-0421.12863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 11/30/2022]
Abstract
Influenza A virus (IAV) PA-X is a critical ribonuclease protein involved in host cell shutoff but its role in modulating the host immune response to IAV infection remains to be addressed. In this study, host cellular proteins that directly interact with PA-X were screened to investigate the biological function of PA-X in the pathogenesis of IAV infection. The protein ankyrin repeat domain 17 (Ankrd17), a positive regulator of inflammatory responses via the retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR) signaling pathway, was identified as a specific PA-X binding partner that preferred PA-X to the PA protein. The N-terminal ankyrin repeats of Ankrd17 are the key domain for the interaction with PA-X rather than PA, which is required for the function of Ankrd17 in elevating the host immune response. Using Ankrd17 knockout and overexpression, we confirmed that PA-X significantly affected the Ankrd17-mediated response to infection in host cells. Our data therefore reveal a novel function for PA-X in the regulation of innate immune pathways via the interaction between PA-X and Ankrd17.
Collapse
Affiliation(s)
- Mai Li
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China.,Guangdong Food and Drug Vocational College, Guangzhou, China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, China
| | - Wenbao Qi
- Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Qing Chang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Ruohong Chen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, China
| | - Danlin Zhen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Ming Liao
- Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, China
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, China
| |
Collapse
|
38
|
Trinh TTT, Duong BT, Nguyen ATV, Tuong HT, Hoang VT, Than DD, Nam S, Sung HW, Yun KJ, Yeo SJ, Park H. Emergence of Novel Reassortant H1N1 Avian Influenza Viruses in Korean Wild Ducks in 2018 and 2019. Viruses 2020; 13:v13010030. [PMID: 33375376 PMCID: PMC7823676 DOI: 10.3390/v13010030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/19/2020] [Accepted: 12/24/2020] [Indexed: 02/08/2023] Open
Abstract
Influenza A virus subtype H1N1 has caused global pandemics like the “Spanish flu” in 1918 and the 2009 H1N1 pandemic several times. H1N1 remains in circulation and survives in multiple animal sources, including wild birds. Surveillance during the winter of 2018–2019 in Korea revealed two H1N1 isolates in samples collected from wild bird feces: KNU18-64 (A/Greater white-fronted goose/South Korea/KNU18-64/2018(H1N1)) and WKU19-4 (A/wild bird/South Korea/WKU19-4/2019(H1N1)). Phylogenetic analysis indicated that M gene of KNU18-64(H1N1) isolate resembles that of the Alaskan avian influenza virus, whereas WKU19-4(H1N1) appears to be closer to the Mongolian virus. Molecular characterization revealed that they harbor the amino acid sequence PSIQRS↓GLF and are low-pathogenicity influenza viruses. In particular, the two isolates harbored three different mutation sites, indicating that they have different virulence characteristics. The mutations in the PB1-F2 and PA protein of WKU19-4(H1N1) indicate increasing polymerase activity. These results corroborate the kinetic growth data for WKU19-4 in MDCK cells: a dramatic increase in the viral titer after 12 h post-inoculation compared with that in the control group H1N1 (CA/04/09(pdm09)). The KNU18-64(H1N1) isolate carries mutations indicating an increase in mammal adaptation; this characterization was confirmed by the animal study in mice. The KNU18-64(H1N1) group showed the presence of viruses in the lungs at days 3 and 6 post-infection, with titers of 2.71 ± 0.16 and 3.71 ± 0.25 log10(TCID50/mL), respectively, whereas the virus was only detected in the WKU19-4(H1N1) group at day 6 post-infection, with a lower titer of 2.75 ± 0.51 log10(TCID50/mL). The present study supports the theory that there is a relationship between Korea and America with regard to reassortment to produce novel viral strains. Therefore, there is a need for increased surveillance of influenza virus circulation in free-flying and wild land-based birds in Korea, particularly with regard to Alaskan and Asian strains.
Collapse
Affiliation(s)
- Thuy-Tien Thi Trinh
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
| | - Bao Tuan Duong
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
| | - Anh Thi Viet Nguyen
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
| | - Hien Thi Tuong
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
| | - Vui Thi Hoang
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
| | - Duong Duc Than
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
| | - SunJeong Nam
- Division of EcoScience, Ewha University, Seoul 03760, Korea;
| | - Haan Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 200-701, Korea;
| | - Ki-Jung Yun
- Department of Pathology, School of Medicine, Wonkwang University, Iksan 570-749, Korea;
| | - Seon-Ju Yeo
- Department of Tropical Medicine and Parasitology, College of Medicine, Seoul National University, Seoul 03080, Korea
- Correspondence: (S.-J.Y.); (H.P.)
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea; (T.-T.T.T.); (B.T.D.); (A.T.V.N.); (H.T.T.); (V.T.H.); (D.D.T.)
- Correspondence: (S.-J.Y.); (H.P.)
| |
Collapse
|
39
|
Sánchez-González R, Ramis A, Nofrarías M, Wali N, Valle R, Pérez M, Perlas A, Majó N. Experimental infection of domestic geese ( Anser anser var. domesticus) with H5N8 Gs/GD and H7N1 highly pathogenic avian influenza viruses. Avian Pathol 2020; 49:642-657. [PMID: 32795171 DOI: 10.1080/03079457.2020.1809635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Prior to the emergence of the Asian-origin H5 Goose/Guangdong/1/96 (Gs/GD) lineage, highly pathogenic avian influenza viruses (HPAIV) had rarely caused high mortalities in domestic geese. In 2016/2017 European epidemics, H5N8 Gs/GD clade 2.3.4.4 Group B produced an unprecedented number of outbreaks in waterfowl holdings. In this study, the pathogenesis of H5N8 HPAIV in comparison with H7N1 HPAIV, and the role of domestic geese in the epidemiology of these viruses, were evaluated. Local and commercial geese (Anser anser var. domesticus) were intranasally inoculated with 105 ELD50 of A/goose/Spain/IA17CR02699/2017 (H5N8) or A/Chicken/Italy/5093/1999 (H7N1) and monitored daily during 15 days. H5N8 was highly virulent to domestic geese, reaching 100% mortality by 10 days post-infection. Systemic microscopic necrotizing lesions associated with widespread AIV-antigen were detected by IHC techniques, the central nervous system being the most severely affected. High viral loads, measured by qRT-PCR, were present in all samples collected: oral and cloacal swabs, plasma tissues, and moderate levels in pool water. Domestic geese were also susceptible to H7N1 infection, as demonstrated by seroconversion and detection of viral RNA in tissues and plasma in some geese, but all lacked clinical signs. Viral shedding was confirmed in only some geese and was restricted to the oral route, but levels were high and still detected at the end of the study. Overall, H7N1 presents a lower lethality and shedding than H5N8 in geese; however, the viral shedding indicates that these species could play a role in the epidemiology of Gs/GD and other lineages of HPAIVs. RESEARCH HIGHLIGHTS H5N8 Gs/GD clade 2.3.4.4 Group B is highly virulent to domestic geese. The severity of H5N8 is associated with multisystemic replication. H7N1 can infect domestic geese but is avirulent to this species. Domestic geese could play a role in the epidemiology of Gs/GD HPAIVs.
Collapse
Affiliation(s)
- R Sánchez-González
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - A Ramis
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - M Nofrarías
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - N Wali
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - R Valle
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - M Pérez
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - A Perlas
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| | - N Majó
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, España
| |
Collapse
|
40
|
Nguyen NM, Sung HW, Yun KJ, Park H, Yeo SJ. 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:v12070774. [PMID: 32709116 PMCID: PMC7411716 DOI: 10.3390/v12070774] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022] Open
Abstract
The complex overlap in waterfowl migratory pathways across the world has established numerous occurrences of genetic reassortment and intercontinental spread of avian influenza virus (AIV) over long distances, thereby calling for huge efforts and targeted surveillance for infection control. During annual surveillance in South Korea in 2018, a novel avian influenza H6N5 (K6) subtype was isolated from the fecal sample of wild bird. Genomic characterization using a phylogenetic tree indicated the K6 virus to be of North American-origin, with partial homology to an H6N5 strain, A/Aix galericulata/South Korea/K17-1638-5/2017 (K17). A monobasic residue at the HA cleavage site and absence of a notable mutation at the HA receptor-binding site suggested the isolate to be of low pathogenicity. However, molecular analysis revealed the E119V mutation in the NA gene and a human host marker mutation E382D in the polymerase acidic (PA) gene, implying their susceptibility to neuraminidase inhibitors and potential infectivity in humans, respectively. For comparison, K6 and K17 were found to be dissimilar for various mutations, such as A274T of PB2, S375N/T of PB1, or V105M of NP, each concerning the increased virulence of K6 in mammalian system. Moreover, kinetic data presented the highest viral titer of this H6N5 isolate at 106.37 log10TCID50 after 48 h of infection, thus proving efficient adaptability for replication in a mammalian system in vitro. The mouse virus challenge study showed insignificant influence on the total body weight, while viral load shedding in lungs peaked at 1.88 ± 0.21 log10 TICD50/mL, six days post infection. The intercontinental transmission of viruses from North America may continuously be present in Korea, thereby providing constant opportunities for virus reassortment with local resident AIVs; these results hint at the increased potential risk of host jumping capabilities of the new isolates. Our findings reinforce the demand for regular surveillance, not only in Korea but also along the flyways in Alaska.
Collapse
Affiliation(s)
- Ngoc Minh Nguyen
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea;
| | - Haan Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 200-701, Korea;
| | - Ki-Jung Yun
- Department of Pathology, School of Medicine, Wonkwang University, Iksan 570-749, Korea;
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea;
- Correspondence: (H.P.); (S.-J.Y.)
| | - Seon-Ju Yeo
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea;
- Correspondence: (H.P.); (S.-J.Y.)
| |
Collapse
|
41
|
Barberis A, Boudaoud A, Gorrill A, Loupias J, Ghram A, Lachheb J, Alloui N, Ducatez MF. Full-length genome sequences of the first H9N2 avian influenza viruses isolated in the Northeast of Algeria. Virol J 2020; 17:108. [PMID: 32680533 PMCID: PMC7366561 DOI: 10.1186/s12985-020-01377-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/30/2020] [Indexed: 11/30/2022] Open
Abstract
Background H9N2 avian influenza viruses (AIV) has a worldwide geographic distribution and affects poultry of different types of production. H9N2 AIV was first reported in the Northeast of Algeria in April 2017, following an outbreak associated with high mortality, in broiler flocks. In the present study, we report full-length genome sequences of AIV H9N2, and the detailed phylogeny and molecular genetic analyses. Methods Ten AIV H9N2 strains, collected in broiler flocks, were amplified in 9-day-old embryonated specific pathogen free (SPF) chicken eggs. Their full-length genomes were successfully sequenced and phylogenetic and molecular characterizations were conducted. Results Phylogenetic analysis showed that the isolates were monophyletic, grouped within the G-1 lineage and were very close to Moroccan and Algerian strains identified in 2016 and 2017, respectively. The low pathogenicity of the strains was confirmed by the sequence motif (335RSSR/GLF341) at the hemagglutinin (HA) cleavage site. An exclusive substitution (T197A) that had not been previously reported for H9N2 viruses; but, conserved in some pandemic H1N1 viruses, was observed. When compared to the G1-like H9N2 prototype, the studied strains showed one less glycosylation site in HA, but 2–3 additional ones in the stalk of the neuraminidase (NA). The HA protein harbored the substitution 234 L, suggesting binding preference to human-like receptors. The NA protein harbored S372A and R403W substitutions, previously detected in H9N2 from Asia and the Middle East, and especially in H2N2 and H3N2 strains that caused human pandemics. Different molecular markers associated with virulence and mammalian infections have been detected in the viral internal proteins. The matrix M2 protein possessed the S31N substitution associated with drug resistance. The non-structural 1 (NS1) protein showed the “GSEV” PDZ ligand (PL) C-terminal motif and no 80–84 deletion. Conclusion Characterized Algerian AIV isolates showed mutations that suggest increased zoonotic potential. Additional studies in animal models are required to investigate the pathogenicity of these H9N2 AIV strains. Monitoring their evolution in both migratory and domestic birds is crucial to prevent transmission to humans. Implementation of adequate biosecurity measures that limit the introduction and the propagation of AIV H9N2 in Algerian poultry farm is crucial.
Collapse
Affiliation(s)
- Abdelheq Barberis
- Centre de Recherche en Biotechnologie, Nouvelle Ville Ali Mendjeli, El Khroub, Algeria. .,LESPA, Département vétérinaire, ISVSA, Université de Batna, Batna, Algeria.
| | - Amine Boudaoud
- LESPA, Département vétérinaire, ISVSA, Université de Batna, Batna, Algeria
| | - Angelina Gorrill
- IHAP, Université de Toulouse, INRAE, ENVT, 23 Chemin des Capelles, 31076, Toulouse cedex, France
| | - Josianne Loupias
- IHAP, Université de Toulouse, INRAE, ENVT, 23 Chemin des Capelles, 31076, Toulouse cedex, France
| | - Abdeljelil Ghram
- Laboratoire d'Epidémiologie et de Microbiologie Vétérinaire, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Jihene Lachheb
- Laboratoire d'Epidémiologie et de Microbiologie Vétérinaire, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Nadir Alloui
- LESPA, Département vétérinaire, ISVSA, Université de Batna, Batna, Algeria
| | - Mariette F Ducatez
- IHAP, Université de Toulouse, INRAE, ENVT, 23 Chemin des Capelles, 31076, Toulouse cedex, France.
| |
Collapse
|
42
|
Arbi M, Souiai O, Rego N, Larbi I, Naya H, Ghram A, Houimel M. Historical origins and zoonotic potential of avian influenza virus H9N2 in Tunisia revealed by Bayesian analysis and molecular characterization. Arch Virol 2020; 165:1527-1540. [PMID: 32335769 DOI: 10.1007/s00705-020-04624-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/24/2020] [Indexed: 01/08/2023]
Abstract
During 2009-2012, several outbreaks of avian influenza virus H9N2 were reported in Tunisian poultry. The circulating strains carried in their hemagglutinins the human-like marker 226L, which is known to be important for avian-to-human viral transmission. To investigate the origins and zoonotic potential of the Tunisian H9N2 viruses, five new isolates were identified during 2012-2016 and their whole genomes were sequenced. Bayesian-based phylogeny showed that the HA, NA, M and NP segments belong to the G1-like lineage. The PB1, PB2, PA and NS segments appeared to have undergone multiple intersubtype reassortments and to be only distantly related to all of the Eurasian lineages (G1-like, Y280-like and Korean-like). The spatiotemporal dynamic of virus spread revealed that the H9N2 virus was transferred to Tunisia from the UAE through Asian and European pathways. As indicated by Bayesian analysis of host traits, ducks and terrestrial birds played an important role in virus transmission to Tunisia. The subtype phylodynamics showed that the history of the PB1 and PB2 segments was marked by intersubtype reassortments with H4N6, H10N4 and H2N2 subtypes. Most of these transitions between locations, hosts and subtypes were statistically supported (BF > 3) and not influenced by sampling bias. Evidence of genetic evolution was observed in the predicted amino acid sequences of the viral proteins of recent Tunisian H9N2 viruses, which were characterized by the acquisition of new mutations involved in virus adaptation to avian and mammalian hosts and amantadine resistance. This study is the first comprehensive analysis of the evolutionary history of Tunisian H9N2 viruses and highlights the zoonotic risk associated with their circulation in poultry, indicating the need for continuous surveillance of their molecular evolution.
Collapse
Affiliation(s)
- Marwa Arbi
- Laboratory of Epidemiology and Veterinary Microbiology, LR19IPT03, Institut Pasteur de Tunis, University Tunis El Manar, 13, Place Pasteur, BP74, 1002, Tunis, Belvedere, Tunisia
| | - Oussema Souiai
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, LR16IPT09, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Natalia Rego
- Bioinformatics Unit, Institut Pasteur de Montevideo, Mataojo 2020, 11400, Montevideo, Uruguay
| | - Imen Larbi
- Laboratory of Epidemiology and Veterinary Microbiology, LR19IPT03, Institut Pasteur de Tunis, University Tunis El Manar, 13, Place Pasteur, BP74, 1002, Tunis, Belvedere, Tunisia
| | - Hugo Naya
- Bioinformatics Unit, Institut Pasteur de Montevideo, Mataojo 2020, 11400, Montevideo, Uruguay
- Departmento de Producción Animal y Pasturas, Facultad de Agronomía, Universidad de la República, Av. Gral. Eugenio Garzón 780, 12900, Montevideo, Uruguay
| | - Abdeljelil Ghram
- Laboratory of Epidemiology and Veterinary Microbiology, LR19IPT03, Institut Pasteur de Tunis, University Tunis El Manar, 13, Place Pasteur, BP74, 1002, Tunis, Belvedere, Tunisia
| | - Mehdi Houimel
- Laboratory of Epidemiology and Veterinary Microbiology, LR19IPT03, Institut Pasteur de Tunis, University Tunis El Manar, 13, Place Pasteur, BP74, 1002, Tunis, Belvedere, Tunisia.
| |
Collapse
|
43
|
Chen Z, Wang Z, Zhao X, Guan Y, Xue Q, Li J, Liu Z, Zhao B, He Z, Huang J, Liao M, Song Y, Jiao P. Pathogenicity of different H5N6 highly pathogenic avian influenza virus strains and host immune responses in chickens. Vet Microbiol 2020; 246:108745. [PMID: 32605756 DOI: 10.1016/j.vetmic.2020.108745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023]
Abstract
The H5N6 highly pathogenic avian influenza virus (HPAIV) has been circulating in China since 2013. In this report, we describe our recent chicken experimental studies investigating the pathogenicity and transmission of four H5N6 HPAIV field strains of different origins (GS39, CK44, DK47 and CK74) and the host immune responses. Four-week-old specific-pathogen-free chickens were inoculated intranasally with one of the four H5N6 HPAIV strains (one strain per group). Among the contact chickens, the GS39 and CK74 strains caused 100 % mortality, the CK44 strain caused 80 % mortality, and the DK47 strain caused 40 % mortality. The viruses were effectively replicated in multiple tissues of the inoculated chickens, in which high viral titers were detected in virus-infected tissues, and significantly upregulated expression of immune-related genes was found in the infected chickens at 24 hpi. The chicken serum antibody levels increased from 5log2 at 7 dpe to 7.67-8log2 at 14 dpe. The major histocompatibility complex molecules were upregulated 21.22- to 32.98-fold in lungs and 5.10- to 18.47-fold in spleens. In summary, H5N6 viruses can replicate within chickens and be effectively transmitted between chickens. Our study contributes to further understanding the pathogenesis of clade 2.3.4.4 H5N6 avian influenza viruses in chickens.
Collapse
Affiliation(s)
- Zuxian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zhenyu Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiya Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yun Guan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qian Xue
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jinrong Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhiting Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bingbing Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhuoliang He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jianni Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yafen Song
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; China Institute of Veterinary Drug Control, Beijing, 100081, China.
| | - Peirong Jiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
44
|
Genetic Characterization of Avian Influenza A (H11N9) Virus Isolated from Mandarin Ducks in South Korea in 2018. Viruses 2020; 12:v12020203. [PMID: 32059510 PMCID: PMC7077279 DOI: 10.3390/v12020203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 01/17/2023] Open
Abstract
In July 2018, a novel avian influenza virus (A/Mandarin duck/South Korea/KNU18-12/2018(H11N9)) was isolated from Mandarin ducks in South Korea. Phylogenetic and molecular analyses were conducted to characterize the genetic origins of the H11N9 strain. Phylogenetic analysis indicated that eight gene segments of strain H11N9 belonged to the Eurasian lineages. Analysis of nucleotide sequence similarity of both the hemagglutinin (HA) and neuraminidase (NA) genes revealed the highest homology with A/duck/Kagoshima/KU57/2014 (H11N9), showing 97.70% and 98.00% nucleotide identities, respectively. Additionally, internal genes showed homology higher than 98% compared to those of other isolates derived from duck and wild birds. Both the polymerase acidic (PA) and polymerase basic 1 (PB1) genes were close to the H5N3 strain isolated in China; whereas, other internal genes were closely related to that of avian influenza virus in Japan. A single basic amino acid at the HA cleavage site (PAIASR↓GLF), the lack of a five-amino acid deletion (residue 69–73) in the stalk region of the NA gene, and E627 in the polymerase basic 2 (PB2) gene indicated that the A/Mandarin duck/South Korea/KNU18-12/2018(H11N9) isolate was a typical low-pathogenicity avian influenza. In vitro viral replication of H11N9 showed a lower titer than H1N1 and higher than H9N2. In mice, H11N9 showed lower adaptation than H1N1. The novel A/Mandarin duck/South Korea/KNU18-12/2018(H11N9) isolate may have resulted from an unknown reassortment through the import of multiple wild birds in Japan and Korea in approximately 2016–2017, evolving to produce a different H11N9 compared to the previous H11N9 in Korea (2016). Further reassortment events of this virus occurred in PB1 and PA in China-derived strains. These results indicate that Japanese- and Chinese-derived avian influenza contributes to the genetic diversity of A/Mandarin duck/South Korea/KNU18-12/2018(H11N9) in Korea.
Collapse
|
45
|
Guan L, Shi J, Kong X, Ma S, Zhang Y, Yin X, He X, Liu L, Suzuki Y, Li C, Deng G, Chen H. H3N2 avian influenza viruses detected in live poultry markets in China bind to human-type receptors and transmit in guinea pigs and ferrets. Emerg Microbes Infect 2020; 8:1280-1290. [PMID: 31495283 PMCID: PMC6746299 DOI: 10.1080/22221751.2019.1660590] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The H3N2 influenza viruses became widespread in humans during the 1968 H3N2 pandemic and have been a major cause of influenza epidemics ever since. Different lineages of H3N2 influenza viruses are also commonly found in animals. If a different lineage of H3N2 virus jumps to humans, a human influenza pandemic could occur with devastating consequences. Here, we studied the genetics, receptor-binding properties, and replication and transmission in mammals of 15 H3N2 avian influenza viruses detected in live poultry markets in China. We found that the H3N2 avian influenza viruses are complicated reassortants with distinct replication phenotypes in mice. Five viruses replicated efficiently in mice and bound to both human-type and avian-type receptors. These viruses transmitted efficiently to direct-contact guinea pigs, and three of them also transmitted among guinea pigs and ferrets via respiratory droplets. Moreover, ferret antiserum induced by human H3N2 viruses did not react with any of the H3N2 avian influenza viruses. Our study demonstrates that the H3N2 avian influenza viruses pose a clear threat to human health and emphasizes the need for continued surveillance and evaluation of the H3N2 influenza viruses circulating in nature.
Collapse
Affiliation(s)
- Lizheng Guan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Jianzhong Shi
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Xingtian Kong
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Shujie Ma
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Yaping Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Xin Yin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Xijun He
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Liling Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Yasuo Suzuki
- College of Life and Health Sciences, Chubu University , Aichi , Japan
| | - Chengjun Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Guohua Deng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS , Harbin , People's Republic of China
| |
Collapse
|
46
|
Lestari, Wibawa H, Lubis EP, Dharmawan R, Rahayu RA, Mulyawan H, Charoenkul K, Nasamran C, Poermadjaja B, Amonsin A. Co-circulation and characterization of HPAI-H5N1 and LPAI-H9N2 recovered from a duck farm, Yogyakarta, Indonesia. Transbound Emerg Dis 2019; 67:994-1007. [PMID: 31770478 DOI: 10.1111/tbed.13434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/30/2019] [Accepted: 11/18/2019] [Indexed: 01/27/2023]
Abstract
In July 2016, an avian influenza outbreak in duck farms in Yogyakarta province was reported to Disease Investigation Center (DIC), Wates, Indonesia, with approximately 1,000 ducks died or culled. In this study, two avian influenza (AI) virus subtypes, A/duck/Bantul/04161291-OR/2016 (H5N1) and A/duck/Bantul/04161291-OP/2016 (H9N2) isolated from ducks in the same farm during an AI outbreak in Bantul district, Yogyakarta province, were sequenced and characterized. Our results showed that H5N1 virus was closely related to the highly pathogenic AI (HPAI) H5N1 of clade 2.3.2.1c, while the H9N2 virus was clustered with LPAI viruses from China, Vietnam and Indonesia H9N2 (CVI lineage). Genetic analysis revealed virulence characteristics for both in avian and in mammalian species. In summary, co-circulation of HPAI-H5N1 of clade 2.3.2.1c and LPAI-H9N2 was identified in a duck farm during an AI outbreak in Yogyakarta province, Indonesia. Our findings raise a concern of the potential risk of the viruses, which could increase viral transmission and/or threat to human health. Routine surveillance of avian influenza viruses should be continuously conducted to understand the dynamic and diversity of the viruses for influenza prevention and control in Indonesia and SEA region.
Collapse
Affiliation(s)
- Lestari
- Department of Veterinary Public Health, Center of Excellence for Emerging and Re-emerging Infectious Diseases in Animals, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Hendra Wibawa
- Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Elly Puspasari Lubis
- Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Rama Dharmawan
- Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Rina Astuti Rahayu
- Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Herdiyanto Mulyawan
- Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Kamonpan Charoenkul
- Department of Veterinary Public Health, Center of Excellence for Emerging and Re-emerging Infectious Diseases in Animals, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Chanakarn Nasamran
- Department of Veterinary Public Health, Center of Excellence for Emerging and Re-emerging Infectious Diseases in Animals, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Bagoes Poermadjaja
- Disease Investigation Center Wates Yogyakarta, Directorate General of Livestock and Animal Health Services, Ministry of Agriculture Indonesia, Yogyakarta, Indonesia
| | - Alongkorn Amonsin
- Department of Veterinary Public Health, Center of Excellence for Emerging and Re-emerging Infectious Diseases in Animals, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
47
|
Sharshov K, Mine J, Sobolev I, Kurskaya O, Dubovitskiy N, Kabilov M, Alikina T, Nakayama M, Tsunekuni R, Derko A, Prokopyeva E, Alekseev A, Shchelkanov M, Druzyaka A, Gadzhiev A, Uchida Y, Shestopalov A, Saito T. Characterization and Phylodynamics of Reassortant H12Nx Viruses in Northern Eurasia. Microorganisms 2019; 7:microorganisms7120643. [PMID: 31816947 PMCID: PMC6956379 DOI: 10.3390/microorganisms7120643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/19/2019] [Accepted: 11/30/2019] [Indexed: 11/16/2022] Open
Abstract
Wild waterfowl birds are known to be the main reservoir for a variety of avian influenza viruses of different subtypes. Some subtypes, such as H2Nx, H8Nx, H12Nx, and H14Nx, occur relatively rarely in nature. During 10-year long-term surveillance, we isolated five rare H12N5 and one H12N2 viruses in three different distinct geographic regions of Northern Eurasia and studied their characteristics. H12N2 from the Far East region was a double reassortant containing hemagglutinin (HA), non-structural (NS) and nucleoprotein (NP) segments of the American lineage and others from the classical Eurasian avian-like lineage. H12N5 viruses contain Eurasian lineage segments. We suggest a phylogeographical scheme for reassortment events associated with geographical groups of aquatic birds and their migration flyways. The H12N2 virus is of particular interest as this subtype has been found in common teal in the Russian Far East region, and it has a strong relation to North American avian influenza virus lineages, clearly showing that viral exchange of segments between the two continents does occur. Our results emphasize the importance of Avian Influenza Virus (AIV) surveillance in Northern Eurasia for the annual screening of virus characteristics, including the genetic constellation of rare virus subtypes, to understand the evolutionary ecology of AIV.
Collapse
Affiliation(s)
- Kirill Sharshov
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
- Correspondence: ; Tel.: +7-960-794-2136; Fax: +7-383-333-6456
| | - Junki Mine
- Division of Transboundary Animal Disease, National Institute of Animal Health, Tsukuba, Ibaraki 305-0856, Japan; (J.M.); (M.N.); (R.T.); (Y.U.); (T.S.)
| | - Ivan Sobolev
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Olga Kurskaya
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Nikita Dubovitskiy
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Marsel Kabilov
- Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia; (M.K.); (T.A.)
| | - Tatiana Alikina
- Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia; (M.K.); (T.A.)
| | - Momoko Nakayama
- Division of Transboundary Animal Disease, National Institute of Animal Health, Tsukuba, Ibaraki 305-0856, Japan; (J.M.); (M.N.); (R.T.); (Y.U.); (T.S.)
| | - Ryota Tsunekuni
- Division of Transboundary Animal Disease, National Institute of Animal Health, Tsukuba, Ibaraki 305-0856, Japan; (J.M.); (M.N.); (R.T.); (Y.U.); (T.S.)
| | - Anastasiya Derko
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Elena Prokopyeva
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Alexander Alekseev
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Michael Shchelkanov
- School of Biomedicine, Far Eastern Federal University, 690091 Vladivostok, Russia;
- Laboratory of Virology, Federal Scientific Center of East Asia Terrestrial Biodiversity, 690022 Vladivostok, Russia
- Laboratory of marine microbiota, National Scientific Center o Marine Biology, 690041 Vladivostok, Russia
| | - Alexey Druzyaka
- Laboratory of behavioral ecology, Institute of Animal Systematics and Ecology, 630091 Novosibirsk, Russia;
| | - Alimurad Gadzhiev
- Department of Ecology, Dagestan State University, 367000 Makhachkala, Russia;
| | - Yuko Uchida
- Division of Transboundary Animal Disease, National Institute of Animal Health, Tsukuba, Ibaraki 305-0856, Japan; (J.M.); (M.N.); (R.T.); (Y.U.); (T.S.)
| | - Alexander Shestopalov
- Department of Experimental Modeling and Pathogenesis of Infectious Diseases, Federal Research Center of Fundamental and Translational Medicine, 630117 Novosibirsk, Russia; (I.S.); (O.K.); (N.D.); (E.P.); (A.A.); (A.S.)
| | - Takehiko Saito
- Division of Transboundary Animal Disease, National Institute of Animal Health, Tsukuba, Ibaraki 305-0856, Japan; (J.M.); (M.N.); (R.T.); (Y.U.); (T.S.)
| |
Collapse
|
48
|
Suttie A, Deng YM, Greenhill AR, Dussart P, Horwood PF, Karlsson EA. Inventory of molecular markers affecting biological characteristics of avian influenza A viruses. Virus Genes 2019; 55:739-768. [PMID: 31428925 PMCID: PMC6831541 DOI: 10.1007/s11262-019-01700-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
Avian influenza viruses (AIVs) circulate globally, spilling over into domestic poultry and causing zoonotic infections in humans. Fortunately, AIVs are not yet capable of causing sustained human-to-human infection; however, AIVs are still a high risk as future pandemic strains, especially if they acquire further mutations that facilitate human infection and/or increase pathogenesis. Molecular characterization of sequencing data for known genetic markers associated with AIV adaptation, transmission, and antiviral resistance allows for fast, efficient assessment of AIV risk. Here we summarize and update the current knowledge on experimentally verified molecular markers involved in AIV pathogenicity, receptor binding, replicative capacity, and transmission in both poultry and mammals with a broad focus to include data available on other AIV subtypes outside of A/H5N1 and A/H7N9.
Collapse
Affiliation(s)
- Annika Suttie
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, 5 Monivong Blvd, PO Box #983, Phnom Penh, Cambodia
- School of Health and Life Sciences, Federation University, Churchill, Australia
- World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yi-Mo Deng
- World Health Organization Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Andrew R Greenhill
- School of Health and Life Sciences, Federation University, Churchill, Australia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, 5 Monivong Blvd, PO Box #983, Phnom Penh, Cambodia
| | - Paul F Horwood
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Erik A Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, 5 Monivong Blvd, PO Box #983, Phnom Penh, Cambodia.
| |
Collapse
|
49
|
Yeo SJ, Than DD, Park HS, Sung HW, Park H. Molecular Characterization of a Novel Avian Influenza A (H2N9) Strain Isolated from Wild Duck in Korea in 2018. Viruses 2019; 11:v11111046. [PMID: 31717636 PMCID: PMC6893532 DOI: 10.3390/v11111046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 01/03/2023] Open
Abstract
A novel avian influenza virus (A/wild duck/Korea/K102/2018) (H2N9) was isolated from wild birds in South Korea in 2018, and phylogenetic and molecular analyses were conducted on complete gene sequences obtained by next-generation sequencing. Phylogenetic analysis indicated that the hemagglutinin (HA) and neuraminidase (NA) genes of the A/wild duck/Korea/K102/2018 (H2N9) virus belonged to the Eurasian countries, whereas other internal genes (polymerase basic protein 1 (PB1), PB2, nucleoprotein (NP), polymerase acidic protein (PA), matrix protein (M), and non-structural protein (NS)) belonged to the East Asian countries. A monobasic amino acid (PQIEPR/GLF) at the HA cleavage site, E627 in the PB2 gene, and no deletion of the stalk region in the NA gene indicated that the A/wild duck/Korea/K102/2018 (H2N9) isolate was a typical low pathogenicity avian influenza (LPAI). Nucleotide sequence similarity analysis of HA revealed that the highest homology (98.34%) is to that of A/duck/Mongolia/482/2015 (H2N3), and amino acid sequence of NA was closely related to that of A/duck/Bangladesh/8987/2010 (H10N9) (96.45%). In contrast, internal genes showed homology higher than 98% compared to those of other isolates derived from duck and wild birds of China or Japan in 2016–2018. The newly isolated A/wild duck/Korea/K102/2018 (H2N9) strain is the first reported avian influenza virus in Korea, and may have evolved from multiple genotypes in wild birds and ducks in Mongolia, China, and Japan.
Collapse
Affiliation(s)
- Seon-Ju Yeo
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (S.-J.Y.); (D.-D.T.)
| | - Duc-Duong Than
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (S.-J.Y.); (D.-D.T.)
| | - Hong-Seog Park
- GnCBio Inc, 4F, Yegan Plaza, 36, Banseok-ro, Yuseong-gu, Daejeon 34069, Korea;
| | - Haan Woo Sung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (H.W.S.); (H.P.)
| | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 54538, Korea; (S.-J.Y.); (D.-D.T.)
- Correspondence: (H.W.S.); (H.P.)
| |
Collapse
|
50
|
Kariithi HM, Welch CN, Ferreira HL, Pusch EA, Ateya LO, Binepal YS, Apopo AA, Dulu TD, Afonso CL, Suarez DL. Genetic characterization and pathogenesis of the first H9N2 low pathogenic avian influenza viruses isolated from chickens in Kenyan live bird markets. INFECTION GENETICS AND EVOLUTION 2019; 78:104074. [PMID: 31634645 DOI: 10.1016/j.meegid.2019.104074] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/11/2019] [Accepted: 10/13/2019] [Indexed: 12/13/2022]
Abstract
Poultry production plays an important role in the economy and livelihoods of rural households in Kenya. As part of a surveillance program, avian influenza virus (AIV)-specific real-time RT-PCR (RRT-PCR) was used to screen 282 oropharyngeal swabs collected from chickens at six live bird markets (LBMs) and 33 backyard poultry farms in Kenya and 8 positive samples were detected. Virus was isolated in eggs from five samples, sequenced, and identified as H9N2 low pathogenic AIV (LPAIV) G1 lineage, with highest nucleotide sequence identity (98.6-99.9%) to a 2017 Ugandan H9N2 isolate. The H9N2 contained molecular markers for mammalian receptor specificity, implying their zoonotic potential. Virus pathogenesis and transmissibility was assessed by inoculating low and medium virus doses of a representative Kenyan H9N2 LPAIV isolate into experimental chickens and exposing them to naïve uninfected chickens at 2 -days post inoculation (dpi). Virus shedding was determined at 2/4/7 dpi and 2/5 days post placement (dpp), and seroconversion determined at 14 dpi/12 dpp. None of the directly-inoculated or contact birds exhibited any mortality or clinical disease signs. All directly-inoculated birds in the low dose group shed virus during the experiment, while only one contact bird shed virus at 2 dpp. Only two directly-inoculated birds that shed high virus titers seroconverted in that group. All birds in the medium dose group shed virus at 4/7 dpi and at 5 dpp, and they all seroconverted at 12/14 dpp. This is the first reported detection of H9N2 LPAIV from Kenya and it was shown to be infectious and transmissible in chickens by direct contact and represents a new disease threat to poultry and potentially to people.
Collapse
Affiliation(s)
- Henry M Kariithi
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O Box 57811, 00200, Kaptagat Road, Loresho, Nairobi, Kenya; Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA.
| | - Catharine N Welch
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA.
| | - Helena L Ferreira
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA; University of Sao Paulo, ZMV- FZEA, Pirassununga 13635900, Brazil.
| | - Elizabeth A Pusch
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA.
| | - Leonard O Ateya
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O Box 57811, 00200, Kaptagat Road, Loresho, Nairobi, Kenya.
| | - Yatinder S Binepal
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O Box 57811, 00200, Kaptagat Road, Loresho, Nairobi, Kenya.
| | - Auleria A Apopo
- Directorate of Veterinary Services, State Department of Livestock, Ministry of Agriculture, Livestock, Fisheries and Irrigation, Private Bag-00625, Nairobi, Kenya.
| | - Thomas D Dulu
- Directorate of Veterinary Services, State Department of Livestock, Ministry of Agriculture, Livestock, Fisheries and Irrigation, Private Bag-00625, Nairobi, Kenya.
| | - Claudio L Afonso
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA.
| | - David L Suarez
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA.
| |
Collapse
|