1
|
Tan KS, Liu J, Andiappan AK, Lew ZZR, He TT, Ong HH, Tay DJW, Aw ZQ, Yi B, Fauzi AM, Yogarajah T, Carmen LCP, Chu JJH, Chow VT, Prabakaran M, Wang DY. Unique immune and other responses of human nasal epithelial cells infected with H5N1 avian influenza virus compared to seasonal human influenza A and B viruses. Emerg Microbes Infect 2025; 14:2484330. [PMID: 40126073 PMCID: PMC11980200 DOI: 10.1080/22221751.2025.2484330] [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: 12/02/2024] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Highly pathogenic avian influenza (HPAI) virus (e.g. H5N1) infects the lower airway to cause severe infections, and constitute a prime candidate for the emergence of disease X. The nasal epithelium is the primary portal of entry for respiratory pathogens, serving as the airway's physical and immune barrier. While HPAI virus predominantly infects the lower airway, not much is known about its interactions with the nasal epithelium. Hence, we sought to elucidate and compare the differential responses of the nasal epithelium against HPAI infection that may contribute to its pathology, and to identify critical response markers. We infected human nasal epithelial cells (hNECs) cultured at the air-liquid interface from multiple healthy donors with clinical isolates of major human seasonal influenza viruses (H1N1, H3N2, influenza B) and HPAI H5N1. The infected cells were subjected to virologic, transcriptomic and secretory protein analyses. While less adapted to infecting the nasal epithelium, HPAI H5N1 elicited unique host responses unlike seasonal influenza. Interestingly, H5N1 infection of hNECs induced responses indicative of subdued antiviral activity (e.g. reduced expression of IFNβ, and inflammasome mediators, IL-1α and IL-1β); decreased wound healing; suppressed re-epithelialization; compromised epithelial barrier integrity; diminished responses to oxidative stress; and increased transmembrane solute and ion carrier gene expression. These unique molecular changes in response to H5N1 infection may represent potential targets for enhancing diagnostic and therapeutic strategies for better surveillance and management of HPAI infection in humans.
Collapse
Affiliation(s)
- Kai Sen Tan
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Liu
- Infectious Diseases Translational Research Programme and Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Zhe Zhang Ryan Lew
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ting Ting He
- Infectious Diseases Translational Research Programme and Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hsiao Hui Ong
- Infectious Diseases Translational Research Programme and Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Douglas Jie Wen Tay
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhen Qin Aw
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bowen Yi
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arfah Mohd Fauzi
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thinesshwary Yogarajah
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Justin Jang Hann Chu
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Collaborative and Translation Unit for HFMD, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Vincent T. Chow
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - De-Yun Wang
- Infectious Diseases Translational Research Programme and Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Ma W, Ren C, Shi L, Meng B, Feng Y, Zhang Y. Isoleucine at position 137 of haemagglutinin acts as a mammalian adaptation marker of H9N2 avian influenza virus. Emerg Microbes Infect 2025; 14:2455597. [PMID: 39817459 PMCID: PMC11789229 DOI: 10.1080/22221751.2025.2455597] [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: 07/22/2024] [Revised: 12/29/2024] [Accepted: 01/14/2025] [Indexed: 01/18/2025]
Abstract
The H9N2 subtype of avian influenza virus (AIV) is widely distributed among poultry and wild birds and is also a threat to humans. During AIV active surveillance in Liaoning province from 2015 to 2016, we identified 10 H9N2 strains exhibiting different lethality to chick embryos. Two representative strains, A/chicken/China/LN07/2016 (CKLN/07) and A/chicken/China/LN17/2016 (CKLN/17), with similar genomic background but different chick embryo lethality, were chosen to evaluate the molecular basis for this difference. A series of reassortants between CKLN/07 and CKLN/17 were generated and their chick embryo lethality was assessed. We found that the isoleucine (I) residue at position 137 (H3 numbering) in the haemagglutinin (HA) was responsible for the chick embryo lethality of the H9N2 virus. Further studies revealed that the threonine (T) to I mutation at HA position 137 enhanced viral replication in vitro and in vivo. Moreover, the HA-T137I substitution in H9N2 avian influenza virus increased the guinea pig transmission efficiency. We also found that the HA-T137I substitution was critical for α2,6-linked sialic acid binding preference and HA activation and stability of H9N2 virus. Our findings demonstrated that HA-137I is a key molecular marker for mammalian adaptation of H9N2 AIV.
Collapse
Affiliation(s)
- Weiwei Ma
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Liaoning Panjin Wetland Ecosystem National Observation and Research Station, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Chenyang Ren
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Liaoning Panjin Wetland Ecosystem National Observation and Research Station, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Lin Shi
- Poultry Diseases Research Laboratory, Liaoning Center for Prevention and Control of Animal Infectious Diseases, Shenyang, People’s Republic of China
| | - Bo Meng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Liaoning Panjin Wetland Ecosystem National Observation and Research Station, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Yali Feng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Liaoning Panjin Wetland Ecosystem National Observation and Research Station, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Ying Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Liaoning Panjin Wetland Ecosystem National Observation and Research Station, Shenyang Agricultural University, Shenyang, People’s Republic of China
| |
Collapse
|
3
|
Ríos Carrasco M, Lin TH, Zhu X, Gabarroca García A, Uslu E, Liang R, Spruit CM, Richard M, Boons GJ, Wilson IA, de Vries RP. The Q226L mutation can convert a highly pathogenic H5 2.3.4.4e virus to bind human-type receptors. Proc Natl Acad Sci U S A 2025; 122:e2419800122. [PMID: 40232794 DOI: 10.1073/pnas.2419800122] [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: 10/01/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
H5Nx viruses continue to wreak havoc in avian and mammalian species worldwide. The virus distinguishes itself by the ability to replicate to high titers and transmit efficiently in a wide variety of hosts in diverse climatic environments. Fortunately, transmission to and between humans is scarce. Yet, if such an event were to occur, it could spark a pandemic as humans are immunologically naïve to H5 viruses. A significant determinant of transmission to and between humans is the ability of the influenza A virus hemagglutinin (HA) protein to shift from an avian-type to a human-type receptor specificity. Here, we demonstrate that a 2016 2.3.4.4e virus HA can convert to human-type receptor binding via a single Q226L mutation, in contrast to a cleavage-modified 2016 2.3.4.4b virus HA. Using glycan arrays, X-ray structural analyses, tissue- and direct glycan binding, we show that L133a Δ and 227Q are vital for this phenotype. Thus, whereas the 2.3.4.4e virus HA only needs a single amino acid mutation, the modified 2016 2.3.4.4b HA was not easily converted to human-type receptor specificity.
Collapse
Affiliation(s)
- María Ríos Carrasco
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
| | - Ting-Hui Lin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Alba Gabarroca García
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
| | - Elif Uslu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
| | - Ruonan Liang
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
| | - Cindy M Spruit
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
| | - Mathilde Richard
- Department of Viroscience, Erasmus University Medical Center, Rotterdam 3000CA, The Netherlands
| | - Geert-Jan Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Robert P de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands
| |
Collapse
|
4
|
Lang Y, Shi L, Roy S, Gupta D, Dai C, Khalid MA, Zhang MZ, Zhang S, Wan XF, Webby R, Ma W. Detection of antibodies against influenza A viruses in cattle. J Virol 2025; 99:e0213824. [PMID: 40130892 PMCID: PMC11998525 DOI: 10.1128/jvi.02138-24] [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: 12/02/2024] [Accepted: 02/15/2025] [Indexed: 03/26/2025] Open
Abstract
Unexpected outbreaks caused by the H5N1 highly pathogenic avian influenza virus (HPAIV) in dairy cows in the United States (US) have raised significant veterinary and public health concerns. When and how the H5N1 HPAIV was introduced into dairy cows and the broader epidemiology of influenza A virus (IAV) infections in cattle in the US remain unclear. Herein, we performed a retrospective study to screen more than 1,700 cattle serum samples collected from different bovine breeds in the US from January 2023 to May 2024 using an enzyme-linked immunosorbent assay (ELISA) targeting the nucleoprotein (NP) to detect IAV infections, and the positive samples were further tested by hemagglutination inhibition (HI) assay. Results showed that 586 of 1,724 samples (33.99%) from 15 US states were seropositive by the NP ELISA assay, including 78 samples collected in 2024 and 508 samples collected in 2023. Moreover, the HI assay revealed that 45 of these ELISA-positive samples were positive to human seasonal H1N1 and H3N2 and swine H3N2 and H1N2 viruses, and some were positive to two or three tested IAVs. Surprisingly, none of these ELISA-positive samples were HI positive for the circulating bovine H5N1 strain. Our results demonstrate that IAVs other than H5N1 can infect cattle, infections are not limited to dairy cows, and that bovine infections with swine and human IAVs have occurred prior to the H5N1 outbreaks. All results highlight the value in monitoring IAV epidemiology in cattle, as the viruses might adapt to cattle and/or reassort with the currently circulating H5N1 HPAIV, increasing risk to humans.IMPORTANCEInfluenza A virus (IAV) is an important zoonotic pathogen that can infect different species. Although cattle were not historically considered vulnerable to IAV infections, an unexpected outbreak caused by H5N1 highly pathogenic avian influenza virus in dairy cows in the United States (US) in early 2024 has raised significant concerns. When and how the virus was introduced into dairy cows and the wider impact of IAV infections in cattle in the US remain unclear. Our retrospective serological screen provided evidence of human and swine H1 and H3 IAV infections in different cattle breeds in addition to dairy cows, although no H5N1 infection was detected. Our results underline the necessity to monitor IAV epidemiology in cattle, as reassortment of IAVs from different species may occur in cattle, generating novel viruses that pose threats to public and animal health.
Collapse
Affiliation(s)
- Yuekun Lang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| | - Lei Shi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| | - Sawrab Roy
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| | - Dipali Gupta
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| | - Chao Dai
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| | - Muhammad Afnan Khalid
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| | - Michael Z. Zhang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Shuping Zhang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Xiu-Feng Wan
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
- Department of Electrical Engineering & Computer Science, College of Engineering, University of Missouri, Columbia, Missouri, USA
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Richard Webby
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Wenjun Ma
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
5
|
Taaffe J, Goldin S, Lambach P, Sparrow E. Global production capacity of seasonal and pandemic influenza vaccines in 2023. Vaccine 2025; 51:126839. [PMID: 39970592 PMCID: PMC11895838 DOI: 10.1016/j.vaccine.2025.126839] [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: 12/10/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Vaccination is a critical part of the response to an influenza pandemic. Future influenza pandemics will likely leverage existing production processes and manufacturing facilities for seasonal influenza to make pandemic vaccines. Therefore, pandemic influenza vaccine response is heavily dependent on seasonal influenza vaccine production capacity. METHODS WHO monitors global vaccine production to inform pandemic preparedness by regularly surveying influenza vaccine manufacturers to estimate both seasonal and potential pandemic vaccine production capacity overall and by region, vaccine type, and manufacturing process. The last survey estimates were for 2019; here, we report updated estimates based on data from the 2023 survey and compare to estimates from previous surveys. RESULTS Our analysis estimates that annual seasonal influenza vaccine production capacity has remained relatively stable since 2019 at 1.53 billion doses and pandemic vaccine capacity at 4.13 and 8.26 billion doses for moderate and best case scenarios, respectively. Over 80 % of seasonal and pandemic vaccine production capacity relies on embryonated eggs, and inactivated influenza virus vaccines comprise the majority of vaccine supply. There is influenza vaccine manufacturing capacity in all WHO regions, except for the African Region, though influenza vaccine production is concentrated in high and upper-middle income countries. The ability to achieve maximum production capacity could be hindered by access to eggs and other ancillary supplies. CONCLUSIONS While influenza vaccine production capacity has been sustained since 2019, significant gaps persist in its distribution, especially in low and lower-middle income countries, and most notably in the African region. This imbalance in production could result in unequal access to vaccines in the event of a pandemic. Strengthening local vaccine manufacturing, promoting seasonal vaccination programmes, and investing in research and development of next-generation influenza vaccines or improved production platforms are essential to improve pandemic preparedness, sustain the influenza vaccine market, and enable more robust local responses.
Collapse
|
6
|
Aranda AJ, Aguilar-Tipacamú G, Perez DR, Bañuelos-Hernandez B, Girgis G, Hernandez-Velasco X, Escorcia-Martinez SM, Castellanos-Huerta I, Petrone-Garcia VM. Emergence, migration and spreading of the high pathogenicity avian influenza virus H5NX of the Gs/Gd lineage into America. J Gen Virol 2025; 106. [PMID: 40279164 DOI: 10.1099/jgv.0.002081] [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] [Indexed: 04/26/2025] Open
Abstract
The high pathogenicity avian influenza virus H5N1, which first emerged in the winter of 2021, has resulted in multiple outbreaks across the American continent through the summer of 2023 and they continue based on early 2025 records, presenting significant challenges for global health and food security. The viruses causing the outbreaks belong to clade 2.3.4.4b, which are descendants of the lineage A/Goose/Guangdong/1/1996 (Gs/Gd) through genetic reassortments with several low pathogenicity avian influenza viruses present in populations of Anseriformes and Charadriiformes orders. This review addresses these issues by thoroughly analysing available epidemiological databases and specialized literature reviews. This project explores the mechanisms behind the resurgence of the H5N1 virus. It provides a comprehensive overview of the origin, timeline and factors contributing to its prevalence among wild bird populations on the American continent.
Collapse
Affiliation(s)
- Alejandro J Aranda
- Maestría en Salud y Producción Animal Sustentable, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Gabriela Aguilar-Tipacamú
- Maestría en Salud y Producción Animal Sustentable, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Licenciatura en Medicina Veterinaria y Zootecnia, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, México
| | - Daniel R Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Bernardo Bañuelos-Hernandez
- Facultad de Veterinaria, Universidad De La Salle Bajío, Avenida Universidad 602, Lomas del Campestre, León, México
| | - George Girgis
- Nevysta Laboratory, Iowa State University Research Park, Ames, Lowa, USA
| | - Xochitl Hernandez-Velasco
- Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia (FMVZ), Universidad Nacional Autónoma de México (UNAM), Cd. de México, México
| | - Socorro M Escorcia-Martinez
- Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia (FMVZ), Universidad Nacional Autónoma de México (UNAM), Cd. de México, México
| | | | - Victor M Petrone-Garcia
- Departamento de Ciencias Pecuarias, Facultad de Estudios Superiores de Cuautitlán (FESC), Universidad Nacional Autónoma de México (UNAM), Cuautitlán, Mexico
| |
Collapse
|
7
|
Liu Y, Deng S, Ren S, Tam RCY, Liu S, Zhang AJ, To KKW, Yuen KY, Chen H, Wang P. Intranasal influenza virus-vectored vaccine offers protection against clade 2.3.4.4b H5N1 infection in small animal models. Nat Commun 2025; 16:3133. [PMID: 40169649 PMCID: PMC11962148 DOI: 10.1038/s41467-025-58504-z] [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: 08/22/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
The highly pathogenic avian influenza (HPAI) H5N1 virus has been endemic in aquatic birds since 1997, causing outbreaks in domestic poultry and occasional human infections worldwide. Recently, the cross-species transmission of a new reassortant variant from clade 2.3.4.4b of H5N1 to cattle in the US has heightened concerns regarding the expansion of host range and potential human infection. As eradicating the H5N1 virus from its reservoir is impossible, it is essential to prepare for a potential pandemic caused by an H5N1 derivative. Utilizing a deleted-NS1 live attenuated influenza viral vector vaccine system (DelNS1 LAIV), a system we have previously used in the development of a COVID-19 vaccine, we have rapidly developed an intranasal vaccine for cattle H5N1 and related clade 2.3.4.4b strains, based on publicly available sequences. Our research demonstrates that a single intranasal immunization can provide effective protection against lethal challenges from HPAI cattle or mink H5N1 variants, offering strong, sustained immunity after two months in female mouse and male hamster models. Immunogenicity analysis reveals that intranasal vaccination with DelNS1 LAIV induces robust neutralizing antibody, mucosal IgA and T cell responses in mice. It is crucial to further evaluate the DelNS1-H5N1 LAIV system to prepare for potential future H5N1 outbreaks in humans.
Collapse
MESH Headings
- Animals
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Administration, Intranasal
- Female
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Mice
- Male
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Cattle
- Humans
- Disease Models, Animal
- Mice, Inbred BALB C
- Antibodies, Viral/immunology
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- Antibodies, Neutralizing/immunology
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shaofeng Deng
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shuang Ren
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Rachel Chun-Yee Tam
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Siwen Liu
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anna Jinxia Zhang
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Pandemic Research Alliance Unit, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Pandemic Research Alliance Unit, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Honglin Chen
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Pui Wang
- State Key Laboratory for Emerging Infectious Diseases and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics Limited, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| |
Collapse
|
8
|
Fu J, Wang P, Wu H, Yang F, Cheng L, Liu F, Yao H, Wu N, Xu L, Wu H. Development of a graphene oxide multilayer quantum dot-based immunochromatographic strip for the ultrasensitive detection of H7 subtype avian influenza viruses. Poult Sci 2025; 104:104924. [PMID: 39985899 PMCID: PMC11904543 DOI: 10.1016/j.psj.2025.104924] [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: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025] Open
Abstract
Since March 2013, the H7N9 subtype of avian influenza virus (AIV) has become an important zoonotic infectious disease, garnering significant global attention because of its potential to affect human health. Establishing a rapid, effective, and sensitive method to detect H7 subtype AIVs is crucial for disease control. In this study, we developed a graphene oxide multilayer quantum dot-based immunochromatographic strip for the ultrasensitive detection of H7 subtype AIVs. The method demonstrated excellent sensitivity, with a limit of detection of 0.063 hemagglutinin units and 0.016 ng/ml for the hemagglutinin protein. The method exhibited remarkable specificity, with no reaction with other subtypes of influenza A virus andno cross-reactivity with other types of avian virus. Additionally, this method exhibited excellent reproducibility, with both inter-group and intra-group variations remaining below 10 %. Preliminary testing on avian clinical samples showed impressive consistency, underscoring the method's reliability. These initial results suggest that this detection approach has significant potential for widespread use in analyzing avian clinical samples, indicating substantial promise for its future application in various diagnostic settings.
Collapse
Affiliation(s)
- Jiamin Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Ping Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Han Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Fan Yang
- Department of Geriatrics, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, PR China
| | - Linfang Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Fumin Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Nanping Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Lihua Xu
- Animal Husbandry and Veterinary Institute, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Haibo Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, and National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| |
Collapse
|
9
|
Zhao C, Huang J, Zhang C, Wang Y, Zhang X, Liu S, Qiang H, Wang H, Zheng H, Zhuang M, Peng Y, Chen F, Zeng X, Chen JL, Ma S. Characteristics of the First Domestic Duck-Origin H12N8 Avian Influenza Virus in China. Int J Mol Sci 2025; 26:2740. [PMID: 40141383 PMCID: PMC11943133 DOI: 10.3390/ijms26062740] [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: 02/11/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
The H12 subtypes of avian influenza viruses (AIVs) are globally prevalent in wild birds, occasionally spilling over into poultry. In this study, we isolated an H12N8 virus from ducks in a live poultry market. Full genomic analysis revealed that the virus bears a single basic amino acid in the cleavage site of the hemagglutinin gene. Phylogenetic analysis revealed that the eight gene segments of the H12N8 virus belong to the Eurasian lineage and the HA gene was clustered with wild bird-originated H12 viruses, with its NP gene showing the highest nucleotide similarity to 2013-like H7N9 viruses. The H12N8 virus replicated effectively in both mammalian and avian cells without prior adaptation. Moreover, the H12N8 virus could infect and replicate in the upper respiratory tract of BALB/c mice without prior adaptation. The H12N8 virus replicated and transmitted inefficiently in both ducks and chickens and hardly triggered high hemagglutination inhibition (HI) antibody titers in the inoculated and contact animals. These results suggest that the wild bird-origin H12N8 virus has reassorted with viruses circulating in domestic poultry, but it inefficiently replicates and transmits in avian hosts. Our findings demonstrate that H12N8 AIV has emerged in domestic poultry, emphasizing the importance of active surveillance of AIVs in both wild and domestic birds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Shujie Ma
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the “Belt and Road”, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (C.Z.); (J.H.); (C.Z.); (Y.W.); (X.Z.); (S.L.); (H.Q.); (H.W.); (H.Z.); (M.Z.); (Y.P.); (F.C.); (X.Z.); (J.-L.C.)
| |
Collapse
|
10
|
Nikolaeva LI, Stuchinskaya MD, Telepenina KP, Shevchenko NG, Kuprianov VV, Krasnoslobodtsev KG, Mukasheva EA, Trushakova SV, Khlopova IN, Kruzhkova IS, Kisteneva LB, Kolobukhina LV, Burtseva EI. Analysis of the association of influenza clinical course with single nucleotide polymorphisms in genes affecting the interferon-λ3 production. Vopr Virusol 2025; 70:25-34. [PMID: 40233334 DOI: 10.36233/0507-4088-271] [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: 10/04/2024] [Accepted: 01/15/2025] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Predisposition to different courses of the infectious process is largely associated with the polymorphisms in human genome, especially in genes encoding proteins of the immune system. In the early stages of influenza infection such components of innate immunity as interferons I (α/β) and III (λ) type play a significant role in limiting virus replication. The aim of the work was to investigate associations of single nucleotide polymorphism in IFNL3 (rs8099917 T/G) and IFNL4 (rs12979860 C/T) genes with different course of influenza, and identify genetic markers of influenza complicated by community-acquired pneumonia. The genes noted above affect the production of interferon-λ3, which is involved in restriction of the viral replication. MATERIALS AND METHODS Samples from 456 patients with mild (n = 150), moderate (n = 173), and severe (n = 133) influenza were studied. The viral RNA was detected by reverse transcription and polymerase chain reaction (RT-PCR). Polymorphisms in IFNL3 (rs8099917 T/G) and IFNL4 (rs12979860 C/T) genes was detected by PCR. Statistical analysis was performed using SNPStats software. RESULTS Patients with the C/T or T/T genotype of IFNL4 gene (rs12979860 C/T) were more likely to have pneumonia than those with the C/C genotype (OR 2.47 (1.31-4.63); p = 0.0044; q = 0.0059). The presence of one T allele increased the risk of developing pneumonia (OR 2.02 (1.05-4.02); p = 0.006; q = 0.008). In the presence of the T/T genotype, the risk increased more than twofold: OR 2.14 (1.31-3.48). Analysis of the SNP of IFNL3 gene (rs8099917 T/G) revealed a weak association of the G allele with pneumonia (OR 1.86 (1.04-3.31); p = 0.03; q = 0.045). CONCLUSION Genetic markers of increased risk of community-acquired pneumonia in influenza include the presence of the T allele in IFNL4 gene (rs12979860 C/T) and, to a lesser extent, the G allele in IFNL3 gene (rs8099917 T/G). Patients carrying these alleles have an increased risk of developing pneumonia, especially in old age.
Collapse
Affiliation(s)
- L I Nikolaeva
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - M D Stuchinskaya
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - K P Telepenina
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - N G Shevchenko
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - V V Kuprianov
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - K G Krasnoslobodtsev
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - E A Mukasheva
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - S V Trushakova
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - I N Khlopova
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - I S Kruzhkova
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - L B Kisteneva
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - L V Kolobukhina
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| | - E I Burtseva
- N.F. Gamaleya National Research Center of Epidemiology and Microbiology of the Ministry of Health of Russia
| |
Collapse
|
11
|
Zhang C, Zhao C, Huang J, Wang Y, Jiang B, Zheng H, Zhuang M, Peng Y, Zhang X, Liu S, Qiang H, Wang H, Zeng X, Guo G, Chen JL, Ma S. Emergence of a novel reassortant H3N3 avian influenza virus with enhanced pathogenicity and transmissibility in chickens in China. Vet Res 2025; 56:56. [PMID: 40069883 PMCID: PMC11899391 DOI: 10.1186/s13567-025-01484-1] [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: 10/10/2024] [Accepted: 01/07/2025] [Indexed: 03/14/2025] Open
Abstract
H3N3 avian influenza viruses (AIVs) are less prevalent in poultry than H3N8 viruses. However, although relatively rare, reassortant H3N3 viruses have been known to appear in both domestic poultry and wild birds. In this study, we isolated the H3N3 virus in chickens sourced from a live poultry market in China. A comprehensive genomic analysis revealed that the virus possessed a single basic amino acid in the cleavage site of the hemagglutinin (HA) gene. Phylogenetic analysis indicated that eight genes in the H3N3 virus belong to the Eurasian lineage. Specifically, the HA and NA genes were clustered with H3N2 and H11N3, respectively, while the internal genes were closely related to the H3N8 and H9N2 viruses. Furthermore, the H3N3 virus exhibited high and moderate stability in thermal and acidic conditions and efficient replication capabilities in mammalian cells. The H3N3 virus demonstrated that it could infect and replicate in the upper and lower respiratory tract of BALB/c mice without prior adaptation, triggering hemagglutination inhibition (HI) antibody titres ranging from 80 to 160; notably, the H3N3 virus replicated vigorously within the chicken respiratory and digestive tracts. The virus also transmitted efficiently and swiftly among chickens through direct contact, leading to higher levels of HI antibodies in both the inoculated and contact birds. These findings suggest that the H3N3 virus may be a novel reassortant originating from viruses circulating in domestic poultry, thus demonstrating an increased pathogenicity and transmissibility in chickens. Our study determines that H3N3 AIV potentially threatens the poultry industry and public health, highlighting the importance of active surveillance of AIVs.
Collapse
Affiliation(s)
- Chunping Zhang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Conghui Zhao
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Jiacheng Huang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Yang Wang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Bo Jiang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Hangyu Zheng
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Mingzhi Zhuang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Yanni Peng
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Xiaoxuan Zhang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Sha Liu
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Haoxi Qiang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Huanhuan Wang
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Xiancheng Zeng
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Guijie Guo
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Ji-Long Chen
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China
| | - Shujie Ma
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, China.
| |
Collapse
|
12
|
Rudometov AP, Litvinova VR, Gudymo AS, Ivanova KI, Rudometova NB, Kisakov DN, Borgoyakova MB, Kisakova LA, Yakovlev VA, Tigeeva EV, Vahitov DI, Makarova KP, Kolosova NP, Ilyicheva TN, Marchenko VY, Sergeev AA, Karpenko LI, Ilyichev AA. Dose-Dependent Effect of DNA Vaccine pVAX-H5 Encoding a Modified Hemagglutinin of Influenza A (H5N8) and Its Cross-Reactivity Against A (H5N1) Influenza Viruses of Clade 2.3.4.4b. Viruses 2025; 17:330. [PMID: 40143259 PMCID: PMC11946855 DOI: 10.3390/v17030330] [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: 12/10/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5 clade 2.3.4.4b viruses are widespread in wild and domestic birds, causing severe economic damage to the global poultry industry. Moreover, viruses of this clade are known to cause infections in mammals, posing a potential pandemic threat. Due to the ongoing evolution and change in the dominant strains of H5 clade 2.3.4.4b, it is important to investigate the cross-reactivity of vaccines in use and under development against clade 2.3.4.4b viruses. In this study, the immunogenicity of the previously developed DNA vaccine encoding a modified hemagglutinin of the influenza A/turkey/Stavropol/320-01/2020 (H5N8) virus, administered by jet injection at doses of 1, 10, 50, 100, and 200 μg, was investigated. The highest titer of specific to recombinant hemagglutinin antibodies was detected in the group of animals injected with 100 µg of DNA vaccine. The cross-reactivity study of sera of animals immunized with 100 µg of DNA vaccine in a microneutralization assay against the strains A/chicken/Astrakhan/321-05/2020 (H5N8), A/chicken/Komi/24-4V/2023 (H5N1), and A/chicken/Khabarovsk/24-1V/2022 (H5N1) showed the formation of cross-neutralizing antibodies. Moreover, the study of protective properties showed that the DNA vaccine protected animals from mortality after infection with A/chicken/Khabarovsk/24-1V/2022 (H5N1) virus.
Collapse
MESH Headings
- Animals
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Cross Reactions
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Influenza A Virus, H5N8 Subtype/immunology
- Influenza A Virus, H5N8 Subtype/genetics
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Influenza in Birds/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Chickens/virology
Collapse
Affiliation(s)
- Andrey P. Rudometov
- Federal Budgetary Research Institution State Research Center of Virology and Biotechnology Vector, Rospotrebnadzor, 630559 Koltsovo, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhou S, Zhang Y, Liu S, Peng C, Shang J, Tian J, Li X, Liu F, Jiang W, Liu H. Pathogenicity of Novel H3 Avian Influenza Viruses in Chickens and Development of a Promising Vaccine. Viruses 2025; 17:288. [PMID: 40143220 PMCID: PMC11946779 DOI: 10.3390/v17030288] [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: 01/06/2025] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 03/28/2025] Open
Abstract
Since 2022, three cases of human infections of novel H3N8 avian influenza viruses (AIVs) have been confirmed in China. Given the potential for significant public health implications, the prompt detection and containment of the virus is particularly important. Comprehensive analyses were conducted of the complete viral gene sequences of five H3 subtype AIVs that were isolated from chickens, pigeons, and geese in live poultry markets in China in 2023. Four strains exhibited a high degree of homology with the H3N8 viruses responsible for human infections in 2022 and 2023. A subsequent study was conducted to investigate the pathogenicity differences among multiple subtypes of the H3 AIVs in chickens. The study revealed that all infected chickens exhibited clinical signs and viral shedding. Notably, two H3N8 viruses, which were highly homologous to human strains, demonstrated significant differences in adaptability to chickens. The goose-derived H3N5 strain displayed high adaptability to chickens and could replicate in multiple organs, with the highest titer in the cloaca. Additionally, a potential vaccine strain, designated CK/NT308/H3N3, was successfully developed that provided complete clinical protection and effectively prevented viral shedding against both H3N3 and H3N8 viruses. In conclusion, CK/NT308/H3N3 presents a promising vaccine candidate.
Collapse
Affiliation(s)
- Shuning Zhou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Yaxin Zhang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Shuo Liu
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Cheng Peng
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Jiajing Shang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Jie Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Taiyuan 030801, China
| | - Xiaoqi Li
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Fuxiao Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Wenming Jiang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Hualei Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| |
Collapse
|
14
|
Yu X, Ni Z, Wang Y, Wang J, Deng G, Shi J, Kong H, Jiang Y, Tian G, Li C, Kawaoka Y, Chen H, Wang J. Claudin-11 plays a pivotal role in the clathrin-mediated endocytosis of influenza A virus. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2856-y. [PMID: 39985647 DOI: 10.1007/s11427-024-2856-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
Identification of host factors that play a key role in viral replication is of great importance for antiviral development. Metabotropic glutamate receptor subtype 2 (mGluR2) is the receptor to trigger clathrin-mediated endocytosis (CME), the major pathway by which influenza virus enters cells. However, other host factors almost certainly involved in the influenza virus CME are largely unknown. Here, we found that the four-transmembrane protein claudin-11 plays an integral part in influenza virus CME. Claudin-11 promotes the dissociation of KCa1.1 (potassium calcium-activated channel subfamily M alpha 1) from mGluR2 and, together with mGluR2, is internalized in virus-containing clathrin-coated pits (CCPs), where it regulates the depolymerization of polymerized F-actin, allowing the CCPs to mature. Importantly, over 60% of claudin-11-silenced mice survived infection with a lethal influenza virus. Our findings advance the understanding of influenza virus infection and provide a promising strategy for the development of host-based antiviral drugs.
Collapse
Affiliation(s)
- Xiaofei Yu
- Key Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Zixin Ni
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yifan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Jingfei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yongping Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Chengjun Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Hualan Chen
- Key Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Jinliang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| |
Collapse
|
15
|
Kawai A, Shimizu T, Tanaka H, Shichinohe S, Anindita J, Hirose M, Kawahara E, Senpuku K, Shimooka M, Quynh Mai LT, Suzuki R, Nogimori T, Yamamoto T, Hirai T, Kato T, Watanabe T, Akita H, Yoshioka Y. Low-inflammatory lipid nanoparticle-based mRNA vaccine elicits protective immunity against H5N1 influenza virus with reduced adverse reactions. Mol Ther 2025; 33:529-547. [PMID: 39690742 PMCID: PMC11852987 DOI: 10.1016/j.ymthe.2024.12.032] [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: 05/31/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024] Open
Abstract
Messenger RNA vaccines based on lipid nanoparticles (mRNA-LNPs) are promising vaccine modalities. However, mRNA-LNP vaccines frequently cause adverse reactions such as swelling and fever in humans, partly due to the inflammatory nature of LNP. Modification of the ionizable lipids used in LNPs is one approach to avoid these adverse reactions. Here, we report the development of mRNA-LNP vaccines with better protective immunity and reduced adverse reactions using LNPs, which contain a disulfide (SS)-cleavable bond and pH-activated lipid-like materials with oleic acid (ssPalmO) as an ionizable lipid (LNPssPalmO). We used mRNA expressing H5N1 subtype high-pathogenicity avian influenza virus-derived hemagglutinin or neuraminidase to generate mRNA-LNP vaccines against H5N1 influenza. Compared with conventional LNPs, mRNA-LNPssPalmO induced comparable antigen-specific antibodies and better interferon-γ (IFN-γ)-producing T helper type 1 responses in mice. Both mRNA-LNPssPalmO and conventional mRNA-LNPs conferred strong protection against homologous H5N1 virus challenge. In addition, mRNA-LNPssPalmO showed better cross-protection against heterologous H5N1 virus challenge compared with conventional mRNA-LNPs. Furthermore, we observed that mRNA-LNPssPalmO induced less-inflammatory responses (e.g., inflammatory cytokine production, vascular hyperpermeability) and fewer adverse reactions (e.g., weight loss, fever) compared with conventional mRNA-LNPs. These results suggest that mRNA-LNPssPalmO would be a safe alternative to conventional vaccines to overcome mRNA-LNP vaccine hesitancy.
Collapse
Affiliation(s)
- Atsushi Kawai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Taro Shimizu
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroki Tanaka
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Shintaro Shichinohe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jessica Anindita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Mika Hirose
- Laboratory for Cryo-EM Structural Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eigo Kawahara
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kota Senpuku
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Makoto Shimooka
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Le Thi Quynh Mai
- Department of Virology, National Institute of Hygiene and Epidemiology, No. 1 Yersin Street, Hanoi 100000, Vietnam
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Toshiro Hirai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kato
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory for Cryo-EM Structural Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tokiko Watanabe
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hidetaka Akita
- Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
16
|
Yang Y, Yang Z, Zhang X, Niu B, Huang Q, Li Y, Yin H, Zhang X, Liao M, Jia W. Rapid detection of Pan-Avian Influenza Virus and H5, H7, H9 subtypes of Avian Influenza Virus using CRISPR/Cas13a and lateral flow assay. Poult Sci 2025; 104:104745. [PMID: 39740498 PMCID: PMC11750554 DOI: 10.1016/j.psj.2024.104745] [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: 11/02/2024] [Revised: 12/18/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025] Open
Abstract
Avian Influenza Virus (AIV) has been prevalent worldwide in recent years, resulting in substantial economic losses in the poultry industry. More importantly, AIV is capable of cross-species transmission among mammals, posing a dormant yet considerable threat to human health and safety. In this study, two rapid detection methods for AIV based on the CRISPR-Cas13a were developed. These methods can identify AIV through the M gene and differentiate the H5, H7, and H9 subtypes via the HA gene. The first method utilizes RT-RAA isothermal amplification of the target sequence in combination with the "collateral effect" of the Cas13a protein. The results are measured using a real-time quantitative PCR instrument, with a Limit of Detection (LOD) as low as 1 copy/μL. The second method combines RT-RAA with Cas13a and a lateral flow assay, allowing results to be visually observed with the naked eye, with a LOD of 10 copies/μL. Both methods demonstrated specificity and sensitivity comparable to or exceeding that of qRT-PCR, suggesting strong potential for clinical application.
Collapse
Affiliation(s)
- Yujia Yang
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhiyi Yang
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xinkui Zhang
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Beibei Niu
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qiuhong Huang
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yan Li
- Engineering Research Center for the Prevention and Control of Animal Original Zoonosis of Fujian Province University, College of Life Science, Longyan University, Longyan, 364012, China
| | - Huifang Yin
- Engineering Research Center for the Prevention and Control of Animal Original Zoonosis of Fujian Province University, College of Life Science, Longyan University, Longyan, 364012, China
| | - Xianpeng Zhang
- Dongguan Key Laboratory of Zoonosis, Dongguan Center for Animal Disease Prevention and Control, Dongguan, 523128, China
| | - Ming Liao
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weixin Jia
- Guangdong Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, National Avian Influenza Para-Reference Laboratory (Guangzhou), College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
17
|
Zhao M, Lei L, Jiang Y, Tian Y, Huang Y, Yang M. Unveiling the Threat of Disease X: Preparing for the Next Global Pandemic. J Med Virol 2025; 97:e70227. [PMID: 39936837 DOI: 10.1002/jmv.70227] [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: 10/15/2024] [Revised: 12/09/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
The term "Disease X", first introduced by the World Health Organization (WHO) in 2018, symbolizes the threat of an unknown pathogen capable of causing a global pandemic. Classified as a "priority pathogens," Disease X stands alongside well-known threats like SARS, Ebola, and ZIKV due to its potential for widespread outbreaks. SARS-CoV-2 is considered the first "Disease X" to fulfill this prediction, demonstrating the devastating impact such pathogens can have. A future pathogen X could pose an even greater threat, with catastrophic consequences. This paper examines the potential origins of such pathogens, drawing lessons from outbreaks like SARS, MERS, and SARS-CoV-2. It also highlights strategic approaches to detect, prevent, and respond effectively to mitigate the risk of future pandemics.
Collapse
Affiliation(s)
- Mengyuan Zhao
- School of Life Science, Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, China
| | - Luping Lei
- Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Yinghan Jiang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, China
| | - Yuxin Tian
- School of Life Science, Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology (BIT), Zhuhai, Guangdong, China
| | - Minghui Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology (BIT), Zhuhai, Guangdong, China
| |
Collapse
|
18
|
Wang G, Jiang L, Wang J, Li Q, Zhang J, Kong F, Yan Y, Wang Y, Deng G, Shi J, Tian G, Zeng X, Liu L, Bu Z, Chen H, Li C. Genome-wide siRNA library screening identifies human host factors that influence the replication of the highly pathogenic H5N1 influenza virus. MLIFE 2025; 4:55-69. [PMID: 40026577 PMCID: PMC11868839 DOI: 10.1002/mlf2.12168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/29/2024] [Accepted: 10/23/2024] [Indexed: 03/05/2025]
Abstract
The global dissemination of H5 avian influenza viruses represents a significant threat to both human and animal health. In this study, we conducted a genome-wide siRNA library screening against the highly pathogenic H5N1 influenza virus, leading us to the identification of 457 cellular cofactors (441 proviral factors and 16 antiviral factors) involved in the virus replication cycle. Gene Ontology term enrichment analysis revealed that the candidate gene data sets were enriched in gene categories associated with mRNA splicing via spliceosome in the biological process, integral component of membrane in the cellular component, and protein binding in the molecular function. Reactome pathway analysis showed that the immune system (up to 63 genes) was the highest enriched pathway. Subsequent comparisons with four previous siRNA library screenings revealed that the overlapping rates of the involved pathways were 8.53%-62.61%, which were significantly higher than those of the common genes (1.85%-6.24%). Together, our genome-wide siRNA library screening unveiled a panorama of host cellular networks engaged in the regulation of highly pathogenic H5N1 influenza virus replication, which may provide potential targets and strategies for developing novel antiviral countermeasures.
Collapse
Affiliation(s)
- Guangwen Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Li Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Jinliang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Qibing Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Jie Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Fandi Kong
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Ya Yan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Yuqin Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Xianying Zeng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Liling Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Zhigao Bu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Chengjun Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| |
Collapse
|
19
|
Kuba Y, Takemae N, Kawato S, Oba K, Taniguchi K, Kageyama T. Evaluation of rapid amplicon-based nanopore sequencing using the latest chemistry for accurate whole genome analysis of influenza A virus in clinical samples. Jpn J Infect Dis 2025:JJID.2024.400. [PMID: 39894472 DOI: 10.7883/yoken.jjid.2024.400] [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: 02/04/2025]
Abstract
MinION sequencing is widely used to sequence influenza A virus (IAV) genomes; however, the accuracy and utility of this approach, using the latest chemistry to obtain whole viral genome sequences directly from clinical samples, remain insufficiently investigated. We evaluated the sequencing accuracy of combining simultaneous multisegment one-step RT-PCR and MinION sequencing using various subtypes of 13 IAV isolates. The latest R10.4.1 chemistry significantly improved sequencing accuracy, achieving ≥99.993% identity with Illumina MiSeq results and reducing the single nucleotide deletion in homopolymer regions. Applying this method to 11 clinical samples enabled rapid subtype identification and the acquisition of eight full-length IAV genomes. In four of these samples, subtype identification of HA and NA was achieved within 20 min after the start of sequencing and a full-length IAV genome was obtained within 7 h after RNA extraction. However, there was concern that cross barcode misassignment during demultiplexing affected data interpretation, particularly for samples with low viral genome copy numbers. This approach can be used for the rapid identification of IAV subtypes and accurate acquisition of full IAV genome sequences from clinical samples, although careful data analysis is required for the multiplex sequencing of clinical samples with low viral genome copy numbers.
Collapse
Affiliation(s)
- Yumani Kuba
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Japan
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, Japan
- Department of Child Medical Health and Development, Mie University Graduate School of Medicine, Japan
| | - Nobuhiro Takemae
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Japan
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, Japan
| | - Satoshi Kawato
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Japan
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, Japan
| | - Kunihiro Oba
- Department of Pediatrics, Showa General Hospital, Japan
- Department of Infection Control and Prevention, Showa General Hospital, Japan
| | - Kiyosu Taniguchi
- Department of Child Medical Health and Development, Mie University Graduate School of Medicine, Japan
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Japan
| | - Tsutomu Kageyama
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Japan
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, Japan
| |
Collapse
|
20
|
Yang K, Nizami S, Hu S, Zou L, Deng H, Xie J, Guo Q, Edwards KM, Dhanasekaran V, Yen HL, Wu J. Genetic diversity of highly pathogenic avian influenza H5N6 and H5N8 viruses in poultry markets in Guangdong, China, 2020-2022. J Virol 2025; 99:e0114524. [PMID: 39629997 PMCID: PMC11784294 DOI: 10.1128/jvi.01145-24] [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: 07/01/2024] [Accepted: 10/30/2024] [Indexed: 02/01/2025] Open
Abstract
H5 highly pathogenic avian influenza (HPAI) viruses of the A/Goose/Guangdong/1/96 (Gs/Gd) lineage continue to evolve and cause outbreaks in domestic poultry and wild birds, with sporadic spillover infections in mammals. The global spread of clade 2.3.4.4b viruses via migratory birds since 2020 has facilitated the introduction of novel reassortants to China, where avian influenza of various subtypes have been epizootic or enzootic among domestic birds. To determine the impact of clade 2.3.4.4b re-introduction on local HPAI dynamics, we analyzed the genetic diversity of H5N6 and H5N8 detected from monthly poultry market surveillance in Guangdong, China, between 2020 and 2022. Our findings reveal that H5N6 viruses clustered in clades 2.3.4.4b and 2.3.4.4h, while H5N8 viruses were exclusively clustered in clade 2.3.4.4b. After 2020, the re-introduced clade 2.3.4.4b viruses replaced the clade 2.3.4.4h viruses detected in 2020. The N6 genes were divided into two clusters, distinguished by an 11 amino acid deletion in the stalk region, while the N8 genes clustered with clade 2.3.4.4 H5N8 viruses circulating among wild birds. Genomic analysis identified 10 transient genotypes. H5N6, which was more prevalently detected, was also clustered into more genotypes than H5N8. Specifically, H5N6 isolates contained genes derived from HPAI H5Nx viruses and low pathogenic avian influenza in China, while the H5N8 isolates contained genes derived from HPAI A(H5N8) 2.3.4.4b and A(H5N1) 2.3.2.1c. No positive selection on amino acid residues associated with mammalian adaptation was found. Our results suggest expanded genetic diversity of H5Nx viruses in China since 2021 with increasing challenges for pandemic preparedness.IMPORTANCESince 2016/2017, clade 2.3.4.4b H5Nx viruses have spread via migratory birds to all continents except Oceania. Here, we evaluated the impact of the re-introduction of clade of 2.3.4.4b on highly pathogenic avian influenza (HPAI) virus genetic diversity in China. Twenty-two H5N6 and H5N8 HPAI isolated from monthly surveillance in two poultry markets in Guangdong between 2020 and 2022 were characterized. Our findings showed that clade 2.3.4.4h, detected in 2020, was replaced by clade 2.3.4.4b in 2021-2022. H5N6 (n = 18) were clustered into more genotypes than H5N8 (n = 4), suggesting that H5N6 may possess better replication fitness in poultry. Conversely, the H5N8 genotypes are largely derived from the clade 2.3.4.4b wild bird isolates. As clade 2.3.4.4b continues to spread via migratory birds, it is anticipated that the genetic diversity of H5N6 viruses circulating in China may continue to expand in the coming years. Continuous efforts in surveillance, genetic analysis, and risk assessment are therefore crucial for pandemic preparedness.
Collapse
Affiliation(s)
- Kang Yang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Sarea Nizami
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Shu Hu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Lirong Zou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Huishi Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jiamin Xie
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Qianfang Guo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Kimberly M. Edwards
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Vijaykrishna Dhanasekaran
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jie Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Wang Z, Tian C, Zhu J, Wang S, Ao X, He Y, Chen H, Liao X, Kong D, Zhou Y, Tai W, Liao M, Fan H. Avian influenza mRNA vaccine encoding hemagglutinin provides complete protection against divergent H5N1 viruses in specific-pathogen-free chickens. J Nanobiotechnology 2025; 23:55. [PMID: 39881325 PMCID: PMC11776166 DOI: 10.1186/s12951-025-03156-w] [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: 05/27/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND The rapid mutation of avian influenza virus (AIV) poses a significant threat to both the poultry industry and public health. Herein, we have successfully developed an mRNA-LNPs candidate vaccine for H5 subtype highly pathogenic avian influenza and evaluated its immunogenicity and protective efficacy. RESULTS In experiments on BALB/c mice, the vaccine candidate elicited strong humoral and a certain cellular immune responses and protected mice from the heterologous AIV challenge. Antibody and splenocyte passive transfer assays in mice suggested that antibodies played a crucial role in providing protection. Experiments involving SPF chickens have revealed that two doses of the 5 µg vaccine candidate in this study provided 100% complete protection against homologous strains, but only 50% complete protection against heterologous strains. Even immunization with two doses of the 15 µg vaccine candidate resulted in 90% complete protection against heterologous strains. To enhance the immune efficacy of the candidate vaccine, we designed 6 sequences with different secondary structures and screened out the candidate sequence with the highest expression (SY2-HA mRNA). Experiments on SPF chickens showed that two doses of 5 µg SY2-HA mRNA-LNP vaccine provided 100% complete protection against homologous and heterologous H5N1 AIV strains. Immunization tests with the SY2-HA mRNA-LNP vaccine were repeated in the SPF chicken model, inducing antibody production levels that are consistent with previous tests and providing 100% complete protection against both homologous and heterologous strains of the virus, indicating that the vaccine has a stable immune efficacy. CONCLUSIONS The vaccine developed in this study provides complete protection against divergent H5N1 AIV strains in chickens, offering a promising approach for the future development of mRNA vaccines against multivalent avian influenza subtypes.
Collapse
Affiliation(s)
- Zhaoyang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Chongyu Tian
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jiahang Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Shiqian Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Xiang Ao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Yanjuan He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Huixin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Xiuying Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Deming Kong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Yongfei Zhou
- Institute of Hemu Biotechnology, Beijing Hemu Biotechnology Co., Ltd, Beijing, 102206, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China.
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China.
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China.
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China.
| |
Collapse
|
22
|
Li B, Raghwani J, Hill SC, François S, Lefrancq N, Liang Y, Wang Z, Dong L, Lemey P, Pybus OG, Tian H. Association of poultry vaccination with interspecies transmission and molecular evolution of H5 subtype avian influenza virus. SCIENCE ADVANCES 2025; 11:eado9140. [PMID: 39841843 PMCID: PMC11753422 DOI: 10.1126/sciadv.ado9140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
The effectiveness of poultry vaccination in preventing the transmission of highly pathogenic avian influenza viruses (AIVs) has been debated, and its impact on wild birds remains uncertain. Here, we reconstruct the movements of H5 subtype AIV lineages among vaccinated poultry, unvaccinated poultry, and wild birds, worldwide, from 1996 to 2023. We find that there is a time lag in viral transmission among different host populations and that movements from wild birds to unvaccinated poultry were more frequent than those from wild birds to vaccinated poultry. Furthermore, our findings suggest that the HA (hemagglutinin) gene of the AIV lineage that circulated predominately in Chinese poultry experienced greater nonsynonymous divergence and adaptive fixation than other lineages. Our results indicate that the epidemiological, ecological, and evolutionary consequences of widespread AIV vaccination in poultry may be linked in complex ways and that much work is needed to better understand how such interventions may affect AIV transmission to, within, and from wild birds.
Collapse
Affiliation(s)
- Bingying Li
- State Key Laboratory of Remote Sensing Science, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Research Center for Respiratory Infectious Diseases, School of National Safety and Emergency Management, Center for Global Change and Public Health, Beijing Normal University, Beijing, China
| | - Jayna Raghwani
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
| | - Sarah C. Hill
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
| | - Sarah François
- Department of Biology, University of Oxford, Oxford, UK
- UMR DGIMI, University of Montpellier, INRAE, Montpellier, France
| | - Noémie Lefrancq
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Yilin Liang
- State Key Laboratory of Remote Sensing Science, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Research Center for Respiratory Infectious Diseases, School of National Safety and Emergency Management, Center for Global Change and Public Health, Beijing Normal University, Beijing, China
| | - Zengmiao Wang
- State Key Laboratory of Remote Sensing Science, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Research Center for Respiratory Infectious Diseases, School of National Safety and Emergency Management, Center for Global Change and Public Health, Beijing Normal University, Beijing, China
| | - Lu Dong
- Ministry of Education Key Laboratory for Biodiversity and Ecological Engineering, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Phillipe Lemey
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
| | - Oliver G. Pybus
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
- Department of Biology, University of Oxford, Oxford, UK
| | - Huaiyu Tian
- State Key Laboratory of Remote Sensing Science, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Research Center for Respiratory Infectious Diseases, School of National Safety and Emergency Management, Center for Global Change and Public Health, Beijing Normal University, Beijing, China
| |
Collapse
|
23
|
Peng P, Shen J, Shi W, Guo J, Wang M, Li W, Yue Z, Sun X, Guan M, Liu L, Xu H, Xie Y, Ren A, Liu M, Liu W, Zhang Z, Xiao Z, Li X. Novel H16N3 avian influenza viruses isolated from migratory gulls in China in 2023. Front Microbiol 2025; 15:1543338. [PMID: 39925884 PMCID: PMC11802517 DOI: 10.3389/fmicb.2024.1543338] [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: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025] Open
Abstract
As a rare subtype of avian influenza virus, H16 viruses are predominant in gulls but rarely found in domestic birds. The low prevalence of H16 viruses has limited our understanding of their epidemiology and evolutionary dynamics. In this study, we isolated three novel H16N3 viruses from migratory gulls in East Asian-Australasian Flyway in eastern China in 2023, which are significantly different from previously identified isolates. To fully understand the epidemiology and genetics characteristics of the global H16 viruses, we compared the host divergence of several rare subtypes and determined that the H13 and H16 subtypes were predominantly pooled into different species of gulls by sharing their internal genes, whereas the waterfowl of Anatidae served as the primary natural reservoirs of the H8, H11, H12, H14, and H15 subtypes. Detailed phylogenetic analysis revealed the evolutionary divergence of globally circulating H16 viruses and their frequent gene reassortment. Furthermore, the gull origin H13 and H16 viruses collectively served as gene donors for the newly emerged highly pathogenic clade 2.3.4.4b H5N1 viruses because the H13/H16-like PA, NP, and NS genes have been introduced into circulating H5N1 viruses since May 2022 in Europe. To date, the H5N1 reassortants containing the H13/H16-like gene segments have been detected in wild and domestic birds and resulted in mammal and human infections. These results improve our knowledge of the ecology and genetics of H16 viruses and emphasize the need for surveillance to monitor the emergence of novel avian influenza viruses in migratory birds.
Collapse
Affiliation(s)
- Peng Peng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Biological Disaster Control and Prevention Center, National Forestry and Grassland Administration, Shenyang, China
| | - Jinyan Shen
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Wenjun Shi
- Technology Center of Qingdao Customs, Qingdao, China
| | - Jing Guo
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Mengjing Wang
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Wenxi Li
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Zhiqin Yue
- Technology Center of Qingdao Customs, Qingdao, China
| | - Xiaohong Sun
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Mengdi Guan
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Lili Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Hongke Xu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Yujiao Xie
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Anran Ren
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Mingfeng Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Wenqiang Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhishu Xiao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuyong Li
- College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| |
Collapse
|
24
|
Martins RP, Marc D, Germon P, Trapp S, Caballero-Posadas I. Influenza A virus in dairy cattle: infection biology and potential mammary gland-targeted vaccines. NPJ Vaccines 2025; 10:8. [PMID: 39805898 PMCID: PMC11730657 DOI: 10.1038/s41541-025-01063-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Influenza, a major "One Health" threat, has gained heightened attention following recent reports of highly pathogenic avian influenza in dairy cattle and cow-to-human transmission in the USA. This review explores general aspects of influenza A virus (IAV) biology, its interactions with mammalian hosts, and discusses the key considerations for developing vaccines to prevent or curtail IAV infection in the bovine mammary gland and its spread through milk.
Collapse
Affiliation(s)
| | - Daniel Marc
- ISP, INRAE, Université de Tours, Nouzilly, France
| | | | - Sascha Trapp
- ISP, INRAE, Université de Tours, Nouzilly, France
| | | |
Collapse
|
25
|
Zhao C, Zhang X, Wang H, Qiang H, Liu S, Zhang C, Huang J, Wang Y, Li P, Chen X, Zhang Z, Ma S. Proteomic Analysis of Differentially Expressed Proteins in A549 Cells Infected with H9N2 Avian Influenza Virus. Int J Mol Sci 2025; 26:657. [PMID: 39859371 PMCID: PMC11765812 DOI: 10.3390/ijms26020657] [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: 11/29/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Influenza A viruses (IAVs) are highly contagious pathogens that cause zoonotic disease with limited availability of antiviral therapies, presenting ongoing challenges to both public health and the livestock industry. Unveiling host proteins that are crucial to the IAV life cycle can help clarify mechanisms of viral replication and identify potential targets for developing alternative host-directed therapies. Using a four-dimensional (4D), label-free methodology coupled with bioinformatics analysis, we analyzed the expression patterns of cellular proteins that changed following H9N2 virus infection. Compared to the control group, the H9N2 infected group displayed 732 differentially expressed proteins (DEPs), with 298 proteins showing upregulation and 434 proteins showing downregulation. Gene Ontology (GO) functional analysis showed that DEPs were catalog in 11 biological processes, three cellular components, and eight molecular functions. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that DEPs were involved in processes including cytokine signaling pathways induced by virus infection and protein digestion and absorption. Proteins including TP53, DDX58, and STAT3 were among the top hub proteins in the protein-protein interaction (PPI) analysis, suggesting that these signaling cascades could be essential for the propagation of IAVs. Furthermore, the host protein SNAPIN was chosen to ascertain the accuracy of expression changes identified through a proteomic analysis. The results indicated that SNAPIN was downregulated following infection with IAVs both in vitro and in vivo, which is consistent with the proteomics results, suggesting that SNAPIN may serve as a key regulatory factor in the viral life cycle of IAVs. Our research delineates an extensive interaction map of IAV infection within the A549 cells, facilitating the discovery of pivotal proteins that contribute to the virus's propagation, potentially offering target candidates to screen for antiviral therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Shujie Ma
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the “Belt and Road”, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (C.Z.); (X.Z.); (H.W.); (H.Q.); (S.L.); (C.Z.); (J.H.); (Y.W.); (P.L.); (X.C.); (Z.Z.)
| |
Collapse
|
26
|
Ríos Carrasco M, Lin TH, Zhu X, García AG, Uslu E, Liang R, Spruit CM, Richard M, Boons GJ, Wilson IA, de Vries RP. The Q226L mutation can convert a highly pathogenic H5 2.3.4.4e virus to bind human-type receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632119. [PMID: 39829928 PMCID: PMC11741302 DOI: 10.1101/2025.01.10.632119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
H5Nx viruses continue to wreak havoc in avian and mammalian species worldwide. The virus distinguishes itself by the ability to replicate to high titers and transmit efficiently in a wide variety of hosts in diverse climatic environments. Fortunately, transmission to and between humans is scarce. Yet, if such an event were to occur, it could spark a pandemic as humans are immunologically naïve to H5 viruses. A significant determinant of transmission to and between humans is the ability of the influenza A virus hemagglutinin (HA) protein to shift from an avian-type to a human-type receptor specificity. Here, we demonstrate that a 2016 2.3.4.4e virus HA can convert to human-type receptor binding via a single Q226L mutation, in contrast to a cleavage-modified 2016 2.3.4.4b virus HA. Using glycan arrays, x-ray structural analyses, tissue- and direct glycan binding, we show that L133aΔ and 227Q are vital for this phenotype. Thus, whereas the 2.3.4.4e virus HA only needs a single amino acid mutation, the modified 2.3.4.4b HA was not easily converted to human-type receptor specificity.
Collapse
Affiliation(s)
- María Ríos Carrasco
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Ting-Hui Lin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alba Gabarroca García
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Elif Uslu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Ruonan Liang
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Cindy M. Spruit
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Mathilde Richard
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Geert-Jan Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA 30602, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robert P. de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| |
Collapse
|
27
|
Wang X, Kong H, Chu B, Yang Q, Lin C, Liu R, Chen C, Gao Y, Wang G, Wang D, Qin C, Ye X, Yu L, Xu X, Jin J, Sun R, Chen H, Wu X, Zhang Z. Identification of a broad-inhibition influenza neuraminidase antibody from pre-existing memory B cells. Cell Host Microbe 2025; 33:151-166.e8. [PMID: 39740671 DOI: 10.1016/j.chom.2024.12.004] [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: 08/02/2024] [Revised: 09/27/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
Identifying broadly reactive B precursor cells and conserved epitopes is crucial for developing a universal flu vaccine. In this study, using influenza neuraminidase (NA) mutant probes, we find that human pre-existing NA-specific memory B cells (MBCs) account for ∼0.25% of total MBCs, which are heterogeneous and dominated by class-unswitched MBCs. In addition, we identify three NA broad-inhibition monoclonal antibodies (mAbs) (BImAbs) that block the activity of NA derived from different influenza strains, including the recent cow H5N1. The cryoelectron microscopy (cryo-EM) structure shows that the BImAb targets the conserved NA enzymatic pocket and a separate epitope in the neighboring NA monomer. Furthermore, the NA BImAbs protect mice from the lethal challenge of the human pandemic H1N1 and H5N1. Our work demonstrates that the NA broad-inhibition precursor MBCs exist in healthy adults and could be targeted by the NA-based universal flu vaccine.
Collapse
Affiliation(s)
- Xin Wang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Bingxin Chu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Qian Yang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Chaohui Lin
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Rui Liu
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Changxu Chen
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Yang Gao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Guojun Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; WHO Collaborating Center for Reference and Research on Influenza, Beijing, China
| | - Chen Qin
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaohua Ye
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Lifei Yu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Xiangfei Xu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jie Jin
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ren Sun
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xudong Wu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Zeli Zhang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Science, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
28
|
Chen T, Gao Y, Chen X, Dong Y, Wang S, Huang Q, Lin S, Wang J, Liao M, Fan H. Self-assembling nanoparticle vaccine elicits a robust protective immune response against avian influenza H5N6 virus in chickens. Int J Biol Macromol 2025; 287:138405. [PMID: 39643188 DOI: 10.1016/j.ijbiomac.2024.138405] [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: 07/06/2024] [Revised: 08/09/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
The continuous circulation and evolution of the H5N6 subtype highly pathogenic avian influenza virus (HPAIV) challenge the development of the global poultry industry and human public health security. To address the potential threat of the H5N6 virus, a secure and efficacious vaccine is urgent. In our research, a self-assembling nanoparticle vaccine presenting the hemagglutinin of the H5N6 AIV was developed based on the ferritin antigen display platform. The results showed that a single-dose vaccination of this nanoparticle vaccine elicited potent hemagglutination inhibition (HI) antibody responses and neutralizing antibody responses in the chickens. Meanwhile, the fused HA-ferritin nanoparticle vaccine induced significantly higher levels of Th1/Th2 immune responses. After a lethal attack with the H5N6 virus, the fused HA-ferritin nanoparticle vaccine conferred chickens with 100 % (10/10) challenge protection. Importantly, the fused HA-ferritin nanoparticle with only 28 hemagglutination units (HAU) provided chickens with immune protection comparable to commercial inactivated vaccines and protected the chickens from severe lung pathological damage. These results in our study support the superiority of ferritin as an antigen display platform and suggest that self-assembled nanoparticle vaccines based on this platform possess the potential as an avian influenza candidate vaccine.
Collapse
Affiliation(s)
- Taoran Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Yinze Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Xingtao Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Yajing Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Shiqian Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Qiao Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Shaorong Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Jiaxin Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China.
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China.
| |
Collapse
|
29
|
Rouygari MR, Mayahi M, Vasfi Marandy M, Boroomand Z. Serological baseline, antibody stability and efficacy of different types of avian influenza (H5) vaccines. J Vet Sci 2025; 26:e4. [PMID: 39901469 PMCID: PMC11799092 DOI: 10.4142/jvs.24045] [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: 02/02/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 02/05/2025] Open
Abstract
IMPORTANCE Evaluating Iran's national highly pathogenic avian influenza (HPAI) control program can inform vaccine selection, optimize immunization duration, guide exit strategies, and assess hemagglutination inhibition (HI) and serum neutralization (SN) methods. OBJECTIVE To establish a serological baseline, assess antibody stability, and compare the efficacy of three HPAI (H5) vaccines. METHODS We analyzed over 9,000 blood samples and 6,420 swabs from approximately 1.5 million birds up to 64 weeks old. HI (β, α), RT-PCR, and SN tests were conducted, with statistical analysis performed using two-way ANOVA. RESULTS The serological baseline (GM titer) using H5N8 antigens from A/Chicken/Iran/162/2016 varied. The Re6+Re8 vaccine produced higher and more stable HI β titers than the H5N3 and baculovirus vaccines. Serum HI α neutralization ability was similar for Re6+Re8 and H5N3 vaccines, both 100 times greater than the baculovirus vaccine. Neutralization indices for H5N3, Re6+Re8, and baculovirus vaccines were 4.7, 4.5, and 4.2 (log2), respectively. CONCLUSIONS AND RELEVANCE After two vaccinations, Re6+Re8 exhibited the most stable HI β antibody response, while H5N3 had the highest neutralization index, surpassing Re6+Re8 by 0.2 and the baculovirus vaccine by 0.5. These findings highlight discrepancies between HI β and SN test results, with SN being a stronger indicator of protective titers due to its in vivo methodology, compared to the in vitro HI assay.
Collapse
Affiliation(s)
- Mohammed Reza Rouygari
- Candidate in Avian Health and Diseases, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 61338-43741, Iran
| | - Mansour Mayahi
- Department of Health and Diseases of Animal, Avian and Aquatic Culture, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 61338-43741, Iran.
| | - Mehdi Vasfi Marandy
- Department of Avian Diseases, Faculty of Veterinary Medicine, Tehran University, Tehran 13117-73833, Iran
| | - Zahra Boroomand
- Department of Health and Diseases of Animal, Avian and Aquatic Culture, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 61338-43741, Iran
| |
Collapse
|
30
|
Luczo JM, Spackman E. Molecular Evolution of the H5 and H7 Highly Pathogenic Avian Influenza Virus Haemagglutinin Cleavage Site Motif. Rev Med Virol 2025; 35:e70012. [PMID: 39730318 DOI: 10.1002/rmv.70012] [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: 07/21/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/29/2024]
Abstract
Avian influenza viruses are ubiquitous in the Anatinae subfamily of aquatic birds and occasionally spill over to poultry. Infection with low pathogenicity avian influenza viruses generally leads to subclinical or mild clinical disease. In contrast, highly pathogenic avian influenza viruses emerge from low pathogenic forms and can cause severe disease associated with extraordinarily high mortality rates. Here, we describe the natural history of avian influenza virus, with a focus on H5Nx and H7Nx subtypes, and the emergence of highly pathogenic forms; we review the biology of AIV; we examine cleavage of haemagglutinin by host cell enzymes with a particular emphasis on the biochemical properties of the proprotein convertases, and trypsin and trypsin-like proteases; we describe mechanisms implicated in the functional evolution of the haemagglutinin cleavage site motif that leads to emergence of HPAIVs; and finally, we discuss the diversity of H5 and H7 haemagglutinin cleavage site sequence motifs. It is crucial to understand the molecular attributes that drive the emergence and evolution of HPAIVs with pandemic potential to inform risk assessments and mitigate the threat of HPAIVs to poultry and human populations.
Collapse
Affiliation(s)
- Jasmina M Luczo
- Australian Animal Health Laboratory, Australian Centre for Disease Preparedness, Commonwealth Scientific and Industrial Research Organisation, East Geelong, Australia
- United States Department of Agriculture, Exotic & Emerging Avian Viral Diseases Research, Southeast Poultry Research Laboratory, United States National Poultry Research Center, Agricultural Research Service, Athens, Georgia, USA
| | - Erica Spackman
- United States Department of Agriculture, Exotic & Emerging Avian Viral Diseases Research, Southeast Poultry Research Laboratory, United States National Poultry Research Center, Agricultural Research Service, Athens, Georgia, USA
| |
Collapse
|
31
|
Kim YW, Jeong S, Yang JH, Tark D, Kim WH, Yang HS, Mun SH, Kang SH, Ko EA, Ko JH. Genetic insights into avian influenza resistance in Jeju Island chickens: the roles of Mx1 and oligoadenylate synthetase-like single nucleotide polymorphisms. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2025; 67:69-85. [PMID: 39974787 PMCID: PMC11833206 DOI: 10.5187/jast.2025.e10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/21/2025]
Abstract
Influenza A virus (FLUAV) causes serious diseases in both poultry and humans. Various host proteins, including Mx1, are considered candidates for avian influenza (AI) resistance. After infecting Jeju Native chicken embryo fibroblasts (CEFs) with three types of AI viruses, we performed gene expression profiling, identified single nucleotide polymorphisms (SNPs) through RNA-sequencing, and confirmed phenotypes showing antiviral activity in vitro. Highly pathogenic AI viruses upregulated FGF2, LYN, and FLT4 and downregulated HGF, ANGPT1, and ROR2, while a low pathogenicity AI upregulated PARK7, RACK1, and DTX3L and downregulated SIRT1, LRRK2, and WAC. However, no virus affected Mx1 expression. Although SNPs in Mx1 could not discriminate antiviral activity alone, the only CEF resistant to H5N6, strain AN4, contained the Mx1 631 R/R genotype and strongly expressed an oligoadenylate synthetase-like (OASL) variant with a unique SNP: c.G880A (p.E294K). Using transfected cell lines, H5N6-infected cells expressing OASL with the c.G880A SNP showed minimal cytopathic effects and the lowest M gene expression. This study confirms that Jeju Native chickens with specific SNP combinations in both Mx1 and OASL showed H5N6 resistance and demonstrates the interplay of genetic factors in host-pathogen dynamics, suggesting a need for integrated analyses of multiple resistance genes to inform AI prevention strategies.
Collapse
Affiliation(s)
- Young-Won Kim
- Department of Physiology, College of
Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Seohyun Jeong
- Department of Physiology, College of
Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Ju-Hee Yang
- Laboratory for Infectious Disease
Prevention, Korea Zoonosis Research Institute, Jeonbuk National
University, Iksan 54531, Korea
| | - Dongseob Tark
- Laboratory for Infectious Disease
Prevention, Korea Zoonosis Research Institute, Jeonbuk National
University, Iksan 54531, Korea
| | - Woo Hyun Kim
- College of Veterinary Medicine &
Institute of Animal Medicine, Gyeongsang National University,
Jinju 52828, Korea
| | | | | | - Sung Hyun Kang
- Chungcheong Regional Center for Disease
Control and Prevention, Korea Disease Control and Prevention
Agency, Daejeon 35208, Korea
| | - Eun-A Ko
- Department of Physiology, School of
Medicine, Jeju National University, Jeju 63243, Korea
| | - Jae-Hong Ko
- Department of Physiology, College of
Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
32
|
Cargnin Faccin F, Perez DR. Pandemic preparedness through vaccine development for avian influenza viruses. Hum Vaccin Immunother 2024; 20:2347019. [PMID: 38807261 PMCID: PMC11141480 DOI: 10.1080/21645515.2024.2347019] [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: 01/30/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Influenza A viruses pose a significant threat to global health, impacting both humans and animals. Zoonotic transmission, particularly from swine and avian species, is the primary source of human influenza outbreaks. Notably, avian influenza viruses of the H5N1, H7N9, and H9N2 subtypes are of pandemic concern through their global spread and sporadic human infections. Preventing and controlling these viruses is critical due to their high threat level. Vaccination remains the most effective strategy for influenza prevention and control in humans, despite varying vaccine efficacy across strains. This review focuses specifically on pandemic preparedness for avian influenza viruses. We delve into vaccines tested in animal models and summarize clinical trials conducted on H5N1, H7N9, and H9N2 vaccines in humans.
Collapse
Affiliation(s)
- Flavio Cargnin Faccin
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Daniel R. Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
33
|
DeDiego ML, Portilla Y, Daviu N, López-García D, Villamayor L, Vázquez-Utrilla P, Mulens-Arias V, Pérez-Yagüe S, Nogales A, Ovejero JG, Gallo-Cordova A, Enjuanes L, Veintemillas-Verdaguer S, Morales MP, Barber DF. Biocompatible Iron Oxide Nanoparticles Display Antiviral Activity Against Two Different Respiratory Viruses in Mice. Int J Nanomedicine 2024; 19:13763-13788. [PMID: 39723174 PMCID: PMC11669338 DOI: 10.2147/ijn.s475323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Background Severe Acute Respiratory syndrome coronavirus 2 (SARS-CoV-2) and Influenza A viruses (IAVs) are among the most important causes of viral respiratory tract infections, causing similar symptoms. IAV and SARS-CoV-2 infections can provoke mild symptoms like fever, cough, sore throat, loss of taste or smell, or they may cause more severe consequences leading to pneumonia, acute respiratory distress syndrome or even death. While treatments for IAV and SARS-CoV-2 infection are available, IAV antivirals often target viral proteins facilitating the emergence of drug-resistant viral variants. Hence, universal treatments against coronaviruses and IAVs are hard to obtain due to genus differences (in the case of coronavirus) or subtypes (in the case of IAV), highlighting the need for novel antiviral therapies. Interestingly, iron oxide nanoparticles (IONPs) with a 10 nm core size and coated with the biocompatible dimercaptosuccinic acid (DMSA: DMSA-IONP-10) display antiviral activity against SARS-CoV-2 in vitro. Methods We analyzed the antiviral activity of DMSA-IONP-10 against SARS-CoV-2 infection in vivo, and against IAV infection in vitro and in vivo. Results DMSA-IONP-10 treatment of mice after SARS-CoV-2 infection impaired virus replication in the lungs and led to a mildly reduced pro-inflammatory cytokine induction after infection, indicating that these IONPs can serve as COVID-19 therapeutic agents. These IONPs also had a prophylactic and therapeutic effect against IAV in tissue cultured cells at non-cytotoxic doses, and a therapeutic effect in IAV-infected-mice, inhibiting viral replication and slightly dampening the inflammatory response after viral infection. As an exacerbated inflammatory response to IAVs and SARS-CoV-2 is detrimental to the host, weakening this response in mice through IONP treatment may reduce disease severity. Interestingly, our data suggest that IONP treatment affects oxidative stress and iron metabolism in cells, which may influence IAV production. Conclusion This study highlights the antiviral activity of DMSA-IONP-10 against important human respiratory viruses.
Collapse
Affiliation(s)
- Marta L DeDiego
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Yadileiny Portilla
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Neus Daviu
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Darío López-García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Laura Villamayor
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Paula Vázquez-Utrilla
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Vladimir Mulens-Arias
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, Madrid, Spain
| | - Jesús G Ovejero
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - Alvaro Gallo-Cordova
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Sabino Veintemillas-Verdaguer
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - M Puerto Morales
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - Domingo F Barber
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
34
|
Verhoeven D, Sponseller BA, Crowe JE, Bangaru S, Webby RJ, Lee BM. Use of equine H3N8 hemagglutinin as a broadly protective influenza vaccine immunogen. NPJ Vaccines 2024; 9:247. [PMID: 39702334 DOI: 10.1038/s41541-024-01037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Development of an efficacious universal influenza vaccines remains a long-sought goal. Current vaccines have shortfalls such as mid/low efficacy and needing yearly strain revisions to account for viral drift/shift. Horses undergo bi-annual vaccines for the H3N8 equine influenza virus, and surveillance of sera from vaccinees demonstrated very broad reactivity and neutralization to many influenza strains. Subsequently, vaccinating mice using the equine A/Kentucky/1/1991 strain or recombinant hemagglutinin (HA) induced similar broadly reactive and neutralizing antibodies to seasonal and high pathogenicity avian influenza strains. Challenge of vaccinated mice protected from lethal virus challenges across H1N1 and H3N2 strains. This protection correlated with neutralizing antibodies to the HA head, esterase, and stem regions. Vaccinated ferrets were also protected after challenge with H1N1 influenza A/07/2009 virus using whole viral or HA. These data suggest that equine H3N8 induces broad protection against multiple influenzas using a unique antigen that diverges from other universal vaccine approaches.
Collapse
Affiliation(s)
- David Verhoeven
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA.
| | - Brett A Sponseller
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA
- Department of Veterinary Clinical Service, Iowa State University, Ames, IA, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt Vaccine Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sandhya Bangaru
- Department of Pediatrics, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude's Children's, Memphis, TN, USA
| | - Brian M Lee
- Department of Chemistry, Coastal Carolina University, Conway, SC, USA
| |
Collapse
|
35
|
Jiao W, Chen Y, Xie Z, Zhao L, Du S, Ma M, Liao M, Dai M. Revealing novel CD8 + T-cell epitopes from the H5N1 avian influenza virus in HBW/B1 haplotype ducks. Vet Res 2024; 55:169. [PMID: 39695865 DOI: 10.1186/s13567-024-01415-6] [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: 06/18/2024] [Accepted: 09/18/2024] [Indexed: 12/20/2024] Open
Abstract
The duck CD8+ T-cell response effectively defends against H5N1 highly pathogenic avian influenza virus (HPAIV) infection, but the recognized peptide is rarely identified. Here, we found that the ratio of CD8+ T cells and the expression of IFN-γ and cytotoxicity-associated genes, including granzyme A/K, perforin and IL2, at 7 days post-infection in peripheral blood mononuclear cells (PBMCs) from B1 haplotype ducks significantly increased in the context of defending against H5N1 AIV infection in vivo. Moreover, similar results were observed in cultured and sorted H5N1 AIV-stimulated duck CD8+ T cells in vitro. Next, we selected 109 epitopes as candidate epitopes on the basis of the MHC-I restriction binding peptide prediction website database and further identified twelve CD8+ T-cell epitopes that significantly increased IFN-γ gene expression after stimulating B1 haplotype duck memory PBMCs. In particular, NP338-346, NP473-481, M2-10, PB1540-548 and PA80-88 were highly conserved in H5N1, H5N6, H5N8, H7N9, and H9N2 AIVs. These findings provide directions for the development of universal T-cell epitope vaccines for AIV in ducks.
Collapse
Affiliation(s)
- Wanlin Jiao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yingyi Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zimin Xie
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Li Zhao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Shanyao Du
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Mulin Ma
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, 510642, China.
| | - Manman Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, 510642, China.
| |
Collapse
|
36
|
Ly H. Highly pathogenic avian influenza H5N1 virus infection of companion animals. Virulence 2024; 15:2289780. [PMID: 38064414 PMCID: PMC10761027 DOI: 10.1080/21505594.2023.2289780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Affiliation(s)
- Hinh Ly
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| |
Collapse
|
37
|
Hastie A, Clarke T, Germain S, Ollinger T, Lese P, Gupta V. Immunogenicity and Safety of AS03-Adjuvanted H7N9 Influenza Vaccine in Adults (18-64 and ≥65 Years): A Phase 1/2, Randomized, Placebo-Controlled Trial. Influenza Other Respir Viruses 2024; 18:e70020. [PMID: 39702896 PMCID: PMC11658966 DOI: 10.1111/irv.70020] [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: 03/28/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Influenza A/Hong Kong/125/2017 (H7N9) virus poses a pandemic risk owing to its evolving nature. This study evaluated the immunogenicity and safety of an AS03-adjuvanted H7N9 vaccine in adults (18-64 years [younger] and ≥65 years [older]). METHODS Participants (younger, n = 418; older, n = 420) were randomized to receive one of six adjuvanted vaccines (hemagglutinin [1.9 μg, 3.75 μg, and 7.5 μg] with AS03A or AS03B) or placebo. The co-primary objectives were to determine whether the adjuvanted vaccines elicit an immune response against the vaccine-homologous virus 21 days after the second vaccine dose and to evaluate the safety of the vaccines. RESULTS H7N9 AS03-adjuvanted vaccines at various doses showed a humoral immune response but failed to meet CBER immunogenicity criteria. However, a trend of increased immune responses was observed with the AS03A adjuvant versus the AS03B adjuvant, particularly in older adults. In both age groups, injection site pain and fatigue occurred more frequently with adjuvanted vaccines. No reported serious adverse events were vaccine-related. CONCLUSIONS This study did not achieve its primary objective at any dose level. The modest immune response to AS03-adjuvanted vaccines, consistent with other studies using similar antigens, highlights the need for continued research for H7N9 pandemic preparedness. TRIAL REGISTRATION NCT04789577 [ClinicalTrials.gov].
Collapse
|
38
|
Shen J, Zhang H, Sun X, Zhang Y, Wang M, Guan M, Liu L, Li W, Xu H, Xie Y, Ren A, Cao F, Liu W, Deng G, Guo J, Li X. Evolution and biological characteristics of H11 avian influenza viruses isolated from migratory birds and pigeons. Emerg Microbes Infect 2024; 13:2398641. [PMID: 39248597 PMCID: PMC11622381 DOI: 10.1080/22221751.2024.2398641] [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: 05/20/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
The emergence of novel avian influenza reassortants in wild birds in recent years is a public health concern. However, the viruses that circulate in migratory birds are not fully understood. In this study, we summarized and categorized global H11 avian influenza viruses and reported that waterfowl and shorebirds are the major reservoirs of the identified H11 viruses. The surveillance data of the 35,749 faecal samples collected from wild bird habitats in eastern China over the past seven years revealed a low prevalence of H11 viruses in birds, with a positive rate of 0.067% (24 isolates). The phylogenetic analysis of the twenty viruses indicated that H11 viruses have undergone complex reassortment with viruses circulating in waterfowl and shorebirds. These tested viruses do not acquire mammalian adaptive mutations in their genomes and preferentially bind to avian-type receptors. Experimental infection studies demonstrated that the two tested H11N9 viruses of wild bird origin replicated and transmitted more efficiently in ducks than in chickens, whereas the pigeon H11N2 virus isolated from a live poultry market was more adapted to replicate in chickens than in ducks. In addition, some H11 isolates replicated efficiently in mice and caused body weight loss but were not lethal. Our study revealed the role of waterfowl and shorebirds in the ecology and evolution of H11 viruses and the potential risk of introducing circulating H11 viruses into ducks or chickens, further emphasizing the importance of avian influenza surveillance at the interface of migratory birds and poultry.
Collapse
Affiliation(s)
- Jinyan Shen
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Hong Zhang
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Xiaohong Sun
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Yaping Zhang
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, State Key Laboratory for Animal Disease Control and Prevention, Harbin, People’s Republic of China
| | - Mengjing Wang
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Mengdi Guan
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Lili Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Wenxi Li
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Hongke Xu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Yujiao Xie
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Anran Ren
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Fengyang Cao
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Wenqiang Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Guohua Deng
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, State Key Laboratory for Animal Disease Control and Prevention, Harbin, People’s Republic of China
| | - Jing Guo
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| | - Xuyong Li
- College of Agriculture and Biology, Liaocheng University, Liaocheng, People’s Republic of China
| |
Collapse
|
39
|
Cui P, Zhuang Y, Zhang Y, Chen L, Chen P, Li J, Feng L, Chen Q, Meng F, Yang H, Jiang Y, Deng G, Shi J, Chen H, Kong H. Does pasteurization inactivate bird flu virus in milk? Emerg Microbes Infect 2024; 13:2364732. [PMID: 38832658 PMCID: PMC11182070 DOI: 10.1080/22221751.2024.2364732] [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: 05/03/2024] [Accepted: 06/02/2024] [Indexed: 06/05/2024]
Abstract
Recently, an outbreak of highly pathogenic avian influenza A (H5N1), which carries the clade 2.3.4.4b hemagglutinin (HA) gene and has been prevalent among North American bird populations since the winter of 2021, was reported in dairy cows in the United States. As of 24 May 2024, the virus has affected 63 dairy herds across nine states and has resulted in two human infections. The virus causes unusual symptoms in dairy cows, including an unexpected drop in milk production, and thick colostrum-like milk. Notably, The US Food and Drug Administration reported that around 20% of tested retail milk samples contained H5N1 viruses, with a higher percentage of positive results from regions with infected cattle herds. Data are scant regarding how effectively pasteurization inactivates the H5N1 virus in milk. Therefore, in this study, we evaluated the thermal stability of the H5 clade 2.3.4.4b viruses, along with one human H3N2 virus and other influenza subtype viruses, including H1, H3, H7, H9, and H10 subtype viruses. We also assessed the effectiveness of pasteurization in inactivating these viruses. We found that the avian H3 virus exhibits the highest thermal stability, whereas the H5N1 viruses that belong to clade 2.3.4.4b display moderate thermal stability. Importantly, our data provide direct evidence that the standard pasteurization methods used by dairy companies are effective in inactivating all tested subtypes of influenza viruses in raw milk. Our findings indicate that thermally pasteurized milk products do not pose a safety risk to consumers.
Collapse
Affiliation(s)
- Pengfei Cui
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yichao Zhuang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yaping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Lei Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Puze Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Jiaqi Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Lulu Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Qin Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Fei Meng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Huanliang Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yongping Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
- Institute of Western Agriculture, CAAS, Changji, People’s Republic of China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| |
Collapse
|
40
|
Xing X, Shi J, Cui P, Yan C, Zhang Y, Zhang Y, Wang C, Chen Y, Zeng X, Tian G, Liu L, Guan Y, Li C, Suzuki Y, Deng G, Chen H. Evolution and biological characterization of H5N1 influenza viruses bearing the clade 2.3.2.1 hemagglutinin gene. Emerg Microbes Infect 2024; 13:2284294. [PMID: 37966008 PMCID: PMC10769554 DOI: 10.1080/22221751.2023.2284294] [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: 08/02/2023] [Accepted: 11/12/2023] [Indexed: 11/16/2023]
Abstract
H5N1 avian influenza viruses bearing the clade 2.3.2.1 hemagglutinin (HA) gene have been widely detected in birds and poultry in several countries. During our routine surveillance, we isolated 28 H5N1 viruses between January 2017 and October 2020. To investigate the genetic relationship of the globally circulating H5N1 viruses and the biological properties of those detected in China, we performed a detailed phylogenic analysis of 274 representative H5N1 strains and analyzed the antigenic properties, receptor-binding preference, and virulence in mice of the H5N1 viruses isolated in China. The phylogenic analysis indicated that the HA genes of the 274 viruses belonged to six subclades, namely clades 2.3.2.1a to 2.3.2.1f; these viruses acquired gene mutations and underwent complicated reassortment to form 58 genotypes, with G43 being the dominant genotype detected in eight Asian and African countries. The 28 H5N1 viruses detected in this study carried the HA of clade 2.3.2.1c (two strains), 2.3.2.1d (three strains), or 2.3.2.1f (23 strains), and formed eight genotypes. These viruses were antigenically well-matched with the H5-Re12 vaccine strain used in China. Animal studies showed that the pathogenicity of the H5N1 viruses ranged from non-lethal to highly lethal in mice. Moreover, the viruses exclusively bound to avian-type receptors and have not acquired the ability to bind to human-type receptors. Our study reveals the overall picture of the evolution of clade 2.3.2.1 H5N1 viruses and provides insights into the control of these viruses.
Collapse
Affiliation(s)
- Xin Xing
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, People’s Republic of China
- Institute of Western Agriculture, CAAS, Changji, People’s Republic of China
| | - Pengfei Cui
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Cheng Yan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yaping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yuancheng Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Congcong Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yuan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Xianying Zeng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Liling Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Yuntao Guan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
- National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Chengjun Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, People’s Republic of China
| | - Yasuo Suzuki
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, Shizuoka, Japan
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, People’s Republic of China
- National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| |
Collapse
|
41
|
Hou Y, Deng G, Cui P, Zeng X, Li B, Wang D, He X, Yan C, Zhang Y, Li J, Ma J, Li Y, Wang X, Tian G, Kong H, Tang L, Suzuki Y, Shi J, Chen H. Evolution of H7N9 highly pathogenic avian influenza virus in the context of vaccination. Emerg Microbes Infect 2024; 13:2343912. [PMID: 38629574 PMCID: PMC11060016 DOI: 10.1080/22221751.2024.2343912] [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: 02/19/2024] [Accepted: 04/11/2024] [Indexed: 05/01/2024]
Abstract
Human infections with the H7N9 influenza virus have been eliminated in China through vaccination of poultry; however, the H7N9 virus has not yet been eradicated from poultry. Carefully analysis of H7N9 viruses in poultry that have sub-optimal immunity may provide a unique opportunity to witness the evolution of highly pathogenic avian influenza virus in the context of vaccination. Between January 2020 and June 2023, we isolated 16 H7N9 viruses from samples we collected during surveillance and samples that were sent to us for disease diagnosis. Genetic analysis indicated that these viruses belonged to a single genotype previously detected in poultry. Antigenic analysis indicated that 12 of the 16 viruses were antigenically close to the H7-Re4 vaccine virus that has been used since January 2022, and the other four viruses showed reduced reactivity with the vaccine. Animal studies indicated that all 16 viruses were nonlethal in mice, and four of six viruses showed reduced virulence in chickens upon intranasally inoculation. Importantly, the H7N9 viruses detected in this study exclusively bound to the avian-type receptors, having lost the capacity to bind to human-type receptors. Our study shows that vaccination slows the evolution of H7N9 virus by preventing its reassortment with other viruses and eliminates a harmful characteristic of H7N9 virus, namely its ability to bind to human-type receptors.
Collapse
Affiliation(s)
- Yujie Hou
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Pengfei Cui
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Xianying Zeng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Bin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Dongxue Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Xinwen He
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Cheng Yan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Yaping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Jiongjie Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Jinming Ma
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
- Institute of Western Agriculture, CAAS, Changji, People's Republic of China
| | - Yanbing Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Xiurong Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Yasuo Suzuki
- Department of Medical Biochemistry, University of Shizuoka School of Pharmaceutical Sciences, Shizuoka, Japan
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
- Institute of Western Agriculture, CAAS, Changji, People's Republic of China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS,Harbin, People’s Republic of China
- National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, CAAS, Harbin, People’s Republic of China
| |
Collapse
|
42
|
Gao F, Wang Q, Qiu C, Luo J, Li X. Pandemic preparedness of effective vaccines for the outbreak of newly H5N1 highly pathogenic avian influenza virus. Virol Sin 2024; 39:981-985. [PMID: 39622294 PMCID: PMC11738761 DOI: 10.1016/j.virs.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
•Analyzed the outbreak situation and viral characteristics of the newly H5N1 highly pathogenic avian influenza (HPAI) virus. •The current approval and research and development of the H5N1 HPAI vaccines were summarized. •Proposed vaccine development approaches against newly H5N1 virus, e.g. adjuvanted vaccine, mRNA vaccine, multivalent vaccine. •Discussed other prevention and control strategies, e.g. poultry vaccination, global surveillance and comprehensive testing.
Collapse
Affiliation(s)
- Feixia Gao
- Shanghai Institute of Biological Products, 200052, Shanghai, China
| | - Qi Wang
- Shanghai Institute of Biological Products, 200052, Shanghai, China
| | - Chenchen Qiu
- Shanghai Institute of Biological Products, 200052, Shanghai, China
| | - Jian Luo
- Shanghai Institute of Biological Products, 200052, Shanghai, China.
| | - Xiuling Li
- Shanghai Institute of Biological Products, 200052, Shanghai, China; State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, Beijing, China.
| |
Collapse
|
43
|
Ameni G, Zewude A, Tulu B, Derara M, Bayissa B, Mohammed T, Degefa BA, Hamad ME, Tibbo M, Barigye R. A Narrative Review on the Pandemic Zoonotic RNA Virus Infections Occurred During the Last 25 Years. J Epidemiol Glob Health 2024; 14:1397-1412. [PMID: 39378018 PMCID: PMC11652441 DOI: 10.1007/s44197-024-00304-7] [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: 06/22/2024] [Accepted: 09/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Pandemic zoonotic RNA virus infections have continued to threaten humans and animals worldwide. The objective of this review was to highlight the epidemiology and socioeconomic impacts of pandemic zoonotic RNA virus infections that occurred between 1997 and 2021. METHODS Literature search was done from Web of Science, PubMed, Google Scholar and Scopus databases, cumulative case fatalities of individual viral infection calculated, and geographic coverage of the pandemics were shown by maps. RESULTS Seven major pandemic zoonotic RNA virus infections occurred from 1997 to 2021 and were presented in three groups: The first group consists of highly pathogenic avian influenza (HPAI-H5N1) and swine-origin influenza (H1N1) viruses with cumulative fatality rates of 53.5% and 0.5% in humans, respectively. Moreover, HPAI-H5N1 infection caused 90-100% death in poultry and economic losses of >$10 billion worldwide. Similarly, H1N1 caused a serious infection in swine and economic losses of 0.5-1.5% of the Gross Domestic Product (GDP) of the affected countries. The second group consists of severe acute respiratory syndrome-associated coronavirus infection (SARS-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Coronavirus disease 2019 (COVID-19) with case fatalities of 9.6%, 34.3% and 2.0%, respectively in humans; but this group only caused mild infections in animals. The third group consists of Ebola and Zika virus infections with case fatalities of 39.5% and 0.02%, respectively in humans but causing only mild infections in animals. CONCLUSION Similar infections are expected in the near future, and hence strict implementation of conventional biosecurity-based measures and development of efficacious vaccines would help minimize the impacts of the next pandemic infection.
Collapse
Affiliation(s)
- Gobena Ameni
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates.
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, PO Box 1176, Addis Ababa, Ethiopia.
| | - Aboma Zewude
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Begna Tulu
- College of Medicine and Health Sciences, Bahir Dar University, P.O. Box 79, Bahir City, Ethiopia
| | - Milky Derara
- Department of Dentistry, College of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia
| | - Berecha Bayissa
- Vaccine Production and Drug Formulation Directorate, National Veterinary Institute, PO Box 35, Debre Zeit, Ethiopia
| | - Temesgen Mohammed
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Berhanu Adenew Degefa
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Mohamed Elfatih Hamad
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Markos Tibbo
- Sub Regional Office for the Gulf-cooperation Council States and Yemen-SNG, Food and Agricultural Organization of the United Nations, Al Qala-id Street, PO Box 62027, Abu Dhabi, United Arab Emirates
| | - Robert Barigye
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| |
Collapse
|
44
|
Zhao Y, Chen P, Hu Y, Liu J, Jiang Y, Zeng X, Deng G, Shi J, Li Y, Tian G, Liu J, Chen H. Recombinant duck enteritis virus bearing the hemagglutinin genes of H5 and H7 influenza viruses is an ideal multivalent live vaccine in ducks. Emerg Microbes Infect 2024; 13:2284301. [PMID: 37966272 PMCID: PMC10769552 DOI: 10.1080/22221751.2023.2284301] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/12/2023] [Indexed: 11/16/2023]
Abstract
Due to the fact that many avian influenza viruses that kill chickens are not lethal to ducks, farmers are reluctant to use avian influenza inactivated vaccines on ducks. Large numbers of unvaccinated ducks play an important role in the transmission of avian influenza viruses from wild birds to domestic poultry, creating a substantial challenge to vaccination strategies for avian influenza control. To solve this problem, we constructed a recombinant duck enteritis virus (DEV), rDEV-dH5/H7, using a live attenuated DEV vaccine strain (vDEV) as a vector. rDEV-dH5/H7 carries the hemagglutinin gene of two H5 viruses [GZ/S4184/17 (H5N6) (clade 2.3.4.4 h) and LN/SD007/17 (H5N1) (clade 2.3.2.1d)] and an H7 virus [GX/SD098/17 (H7N9)]. These three hemagglutinin genes were stably inherited in rDEV-dH5/H7 and expressed in rDEV-dH5/H7-infected cells. Animal studies revealed that rDEV-dH5/H7 and vDEV induced similar neutralizing antibody responses and protection against lethal DEV challenge. Importantly, rDEV-dH5/H7 induced strong and long-lasting hemagglutinin inhibition antibodies against different H5 and H7 viruses and provided complete protection against challenges with homologous and heterologous highly pathogenic H5 and H7 influenza viruses in ducks. Our study shows that rDEV-dH5/H7 could serve as an ideal live attenuated vaccine to protect ducks against infection with lethal DEV and highly pathogenic avian influenza viruses.
Collapse
Affiliation(s)
- Yubo Zhao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Pucheng Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Yuzhen Hu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Jing Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Yongping Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Xianying Zeng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Yanbing Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Jinxiong Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
- National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
- National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| |
Collapse
|
45
|
Plaza PI, Lambertucci SA. Unsustainable production patterns and disease emergence: The paradigmatic case of Highly Pathogenic Avian Influenza H5N1. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175389. [PMID: 39134272 DOI: 10.1016/j.scitotenv.2024.175389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024]
Abstract
Current food production systems are causing severe environmental damage, including the emergence of dangerous pathogens that put humans and wildlife at risk. Several dangerous pathogens (e.g., the 2009 A(H1N1) Influenza Virus, Nipah virus) have emerged associated with the dominant intensive food production systems. In this article, we use the case of the emergence and spillover of the Highly Pathogenic Avian Influenza virus H5N1 (hereafter, H5N1) to illustrate how intensive food production methods provide a breeding ground for dangerous pathogens. We also discuss how emerging pathogens, such as H5N1, may affect not only ecosystem health but also human well-being and the economy. The current H5N1 panzootic (2020-2024) is producing a catastrophic impact: the millions of domestic birds affected by this virus have led to significant economic losses globally, and wild birds and mammals have suffered alarming mortalities, with the associated loss of their material and non-material ecosystem services. Transformative actions are required to reduce the emergence and impact of pathogens such as H5N1; we particularly need to reconsider the ways we are producing food. Governments should redirect funds to the promotion of alternative production systems that reduce the risk of new emerging pathogens and produce environmentally healthy food. These systems need to have a positive relationship with nature rather than being systems based on business as usual to the detriment of the environment. Sustainable food production systems may save many lives, economies, and biodiversity, together with the ecosystem services species provide.
Collapse
Affiliation(s)
- Pablo I Plaza
- Grupo de Investigaciones en Biología de la Conservación, Laboratorio Ecotono, INIBIOMA, Universidad Nacional del Comahue - CONICET, Quintral 1250 (R8400FRF), San Carlos de Bariloche, Argentina.
| | - Sergio A Lambertucci
- Grupo de Investigaciones en Biología de la Conservación, Laboratorio Ecotono, INIBIOMA, Universidad Nacional del Comahue - CONICET, Quintral 1250 (R8400FRF), San Carlos de Bariloche, Argentina
| |
Collapse
|
46
|
Marchenko VY, Panova AS, Kolosova NP, Gudymo AS, Svyatchenko SV, Danilenko AV, Vasiltsova NN, Egorova ML, Onkhonova GS, Zhestkov PD, Zinyakov NG, Andreychuk DB, Chvala IA, Kosenko MN, Moiseeva AA, Boldyrev ND, Shadrinova KN, Perfilieva ON, Ryzhikov AB. Characterization of H5N1 avian influenza virus isolated from bird in Russia with the E627K mutation in the PB2 protein. Sci Rep 2024; 14:26490. [PMID: 39489822 PMCID: PMC11532466 DOI: 10.1038/s41598-024-78175-y] [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: 07/31/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
Currently A(H5Nx) avian influenza viruses are globally widespread and continue to evolve. Since their emergence in 2020 novel highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b reassortant viruses have become predominant in the world and caused multiple infections in mammals. It was shown that some of A(H5N1) viruses mostly isolated from mammals contain an E627K mutation in the PB2 protein which can lead to adaptation of influenza viruses to mammalian cells. In 2023 in Russia we have isolated two highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b viruses from birds one of which contained an E627K mutation in the PB2 protein. This virus had increased virulence in mice. Limited airborne transmission of the virus with the PB2-E627K mutation was observed between ferrets, in which infectious virus was detected in the nasal washings of the three of the twelve recipient ferrets, and clinical symptoms of the disease were observed in one case. Both viruses showed dominant binding to avian-type sialoside receptors, which was most likely the reason for the limited transmissibility. Thus, this study indicates a possible limited increase in the pandemic potential of A(H5N1) 2.3.4.4b viruses and highlights the importance of continuous avian influenza surveillance for pandemic preparedness and response.
Collapse
Affiliation(s)
- Vasiliy Yu Marchenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia.
| | - Anastasia S Panova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Natalia P Kolosova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Andrey S Gudymo
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Svetlana V Svyatchenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Alexey V Danilenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Natalia N Vasiltsova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Marina L Egorova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Galina S Onkhonova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Pavel D Zhestkov
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Nikolay G Zinyakov
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Dmitriy B Andreychuk
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Ilya A Chvala
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Maksim N Kosenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Anastasia A Moiseeva
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Nikita D Boldyrev
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Kiunnei N Shadrinova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Olga N Perfilieva
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Alexander B Ryzhikov
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| |
Collapse
|
47
|
Zhao L, Li S, Deng L, Zhang Y, Jiang C, Wei Y, Xia J, Ping J. Host-specific SRSF7 regulates polymerase activity and replication of influenza A virus. Microbes Infect 2024; 26:105401. [PMID: 39134172 DOI: 10.1016/j.micinf.2024.105401] [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: 02/07/2024] [Revised: 05/19/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Avian influenza viruses crossing the host barrier to infect humans have caused great panic in human society and seriously threatened public health. Herein, we revealed that knockdown of SRSF7 significantly down-regulated influenza virus titers and viral protein expression. We further observed for the first time that human SRSF7, but not avian SRSF7, significantly inhibited polymerase activity (PB2627E). Molecular mapping demonstrated that amino acids 206 to 228 of human SRSF7 play a decisive role in regulating the polymerase activity, which contains the amino acid motif absent in avian SRSF7. Importantly, our results illustrated that the PB2627K-encoding influenza virus induces SRSF7 protein degradation more strongly via the lysosome pathway and not via the proteasome pathway. Functional enrichment analysis of SRSF7-related KEGG pathways indicated that SRSF7 is closely related to cell growth and death. Lastly, our results showed that knocking down SRSF7 interferes with normal polymerase activity. Taken together, our results advance our understanding of interspecies transmission and our findings point out new targets for the development of drugs preventing or treating influenza virus infection.
Collapse
Affiliation(s)
- Lingcai Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Shengmin Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Lulu Deng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Yijia Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Chenfeng Jiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Yurong Wei
- Xinjiang Key Laboratory of Animal Infectious Diseases, Institute of Veterinary Medicine, Xinjiang Academy of Animal Sciences, Urumqi, 830013, China.
| | - Jun Xia
- Key Laboratory of Herbivore Disease Prevention and Control, (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, China.
| | - Jihui Ping
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
48
|
Nakhaie M, Rukerd MRZ, Shahpar A, Pardeshenas M, Khoshnazar SM, Khazaeli M, Bashash D, Nezhad NZ, Charostad J. A Closer Look at the Avian Influenza Virus H7N9: A Calm before the Storm? J Med Virol 2024; 96:e70090. [PMID: 39601174 DOI: 10.1002/jmv.70090] [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: 05/27/2024] [Revised: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The avian influenza A (H7N9) virus, which circulates in wild birds and poultry, has been a major concern for public health since it was first discovered in China in 2013 due to its demonstrated ability to infect humans, causing severe respiratory illness with high mortality rates. According to the World Health Organization (WHO), a total of 1568 human infections with 616 fatal cases caused by novel H7N9 viruses have been reported in China from early 2013 to January 2024. This manuscript provides a comprehensive review of the virology, evolutionary patterns, and pandemic potential of H7N9. The H7N9 virus exhibits a complex reassortment history, receiving genes from H9N2 and other avian influenza viruses. The presence of certain molecular markers, such as mutations in the hemagglutinin and polymerase basic protein 2, enhances the virus's adaptability to human hosts. The virus activates innate immune responses through pattern recognition receptors, leading to cytokine production and inflammation. Clinical manifestations range from mild to severe, with complications including pneumonia, acute respiratory distress syndrome, and multiorgan failure. Diagnosis relies on molecular assays such as reverse transcription-polymerase chain reaction. The increasing frequency of human infections, along with the virus's ability to bind to human receptors and cause severe disease, highlights its pandemic potential. Continued surveillance, vaccine development, and public health measures are crucial to limit the risk posed by H7N9. Understanding the virus's ecology, transmission dynamics, and pathogenesis is essential for developing effective prevention and control strategies.
Collapse
Affiliation(s)
- Mohsen Nakhaie
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Rezaei Zadeh Rukerd
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amirhossein Shahpar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Pardeshenas
- Department of Microbiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mana Khazaeli
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Zeinali Nezhad
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Javad Charostad
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
49
|
Sreenivasan CC, Li F, Wang D. Emerging Threats of Highly Pathogenic Avian Influenza A (H5N1) in US Dairy Cattle: Understanding Cross-Species Transmission Dynamics in Mammalian Hosts. Viruses 2024; 16:1703. [PMID: 39599818 PMCID: PMC11598956 DOI: 10.3390/v16111703] [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: 08/19/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
The rapid geographic spread of the highly pathogenic avian influenza (HPAI) A(H5N1) virus in poultry, wild birds, and other mammalian hosts, including humans, raises significant health concerns globally. The recent emergence of HPAI A(H5N1) in agricultural animals such as cattle and goats indicates the ability of the virus to breach unconventional host interfaces, further expanding the host range. Among the four influenza types-A, B, C, and D, cattle are most susceptible to influenza D infection and serve as a reservoir for this seven-segmented influenza virus. It is generally thought that bovines are not hosts for other types of influenza viruses, including type A. However, this long-standing viewpoint has been challenged by the recent outbreaks of HPAI A(H5N1) in dairy cows in the United States. To date, HPAI A(H5N1) has spread into fourteen states, affecting 299 dairy herds and causing clinical symptoms such as reduced appetite, fever, and a sudden drop in milk production. Infected cows can also transmit the disease through raw milk. This review article describes the current epidemiological landscape of HPAI A(H5N1) in US dairy cows and its interspecies transmission events in other mammalian hosts reported across the globe. The review also discusses the viral determinants of tropism, host range, adaptative mutations of HPAI A(H5N1) in various mammalian hosts with natural and experimental infections, and vaccination strategies. Finally, it summarizes some immediate questions that need to be addressed for a better understanding of the infection biology, transmission, and immune response of HPAI A(H5N1) in bovines.
Collapse
Affiliation(s)
| | - Feng Li
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, KY 40546, USA;
| | - Dan Wang
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, KY 40546, USA;
| |
Collapse
|
50
|
Deng M, Chen S, Wu J, Su L, Xu Z, Jiang C, Sheng L, Yang X, Zeng L, Wang J, Dai W. Exploring the anti-inflammatory and immune regulatory effects of Taohe Chengqi decoction in sepsis-induced lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118404. [PMID: 38824977 DOI: 10.1016/j.jep.2024.118404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sepsis presents complex pathophysiological challenges. Taohe Chengqi Decoction (THCQ), a traditional Chinese medicine, offers potential in managing sepsis-related complications, though its exact mechanisms are not fully understood. AIM OF THE STUDY This research aimed to assess the therapeutic efficacy and underlying mechanisms of THCQ on sepsis-induced lung injury. MATERIALS AND METHODS The study began with validating THCQ's anti-inflammatory effects through in vitro and in vivo experiments. Network pharmacology was employed for mechanistic exploration, incorporating GO, KEGG, and PPI analyses of targets. Hub gene-immune cell correlations were assessed using CIBERSORT, with further scrutiny at clinical and single-cell levels. Molecular docking explored THCQ's drug-gene interactions, culminating in qPCR and WB validations of hub gene expressions in sepsis and post-THCQ treatment scenarios. RESULTS THCQ demonstrated efficacy in modulating inflammatory responses in sepsis, identified through network pharmacology. Key genes like MAPK14, MAPK3, MMP9, STAT3, LYN, AKT1, PTPN11, and HSP90AA1 emerged as central targets. Molecular docking revealed interactions between these genes and THCQ components. qPCR results showed significant modulation of these genes, indicating THCQ's potential in reducing inflammation and regulating immune responses in sepsis. CONCLUSION This study sheds light on THCQ's anti-inflammatory and immune regulatory mechanisms in sepsis, providing a foundation for further research and potential clinical application.
Collapse
Affiliation(s)
- Mingtao Deng
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Siqi Chen
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Jian Wu
- Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Liling Su
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Zijin Xu
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Changrun Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Lei Sheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Xinyi Yang
- Department of Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwaizheng Street, Dong Lake District, Nanchang, Jiangxi Province, 330000, People's Republic of China
| | - Long Zeng
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Jingwei Wang
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Wei Dai
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Clinical Medicine, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China.
| |
Collapse
|