1
|
Krammer F, Hermann E, Rasmussen AL. Highly pathogenic avian influenza H5N1: history, current situation, and outlook. J Virol 2025:e0220924. [PMID: 40145745 DOI: 10.1128/jvi.02209-24] [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: 03/28/2025] Open
Abstract
The H5N1 avian panzootic has resulted in cross-species transmission to birds and mammals, causing outbreaks in wildlife, poultry, and US dairy cattle with a range of host-dependent pathogenic outcomes. Although no human-to-human transmission has been observed, the rising number of zoonotic human cases creates opportunities for adaptive mutation or reassortment. This Gem explores the history, evolution, virology, and epidemiology of clade 2.3.4.4b H5N1 relative to its pandemic potential. Pandemic risk reduction measures are urgently required.
Collapse
Affiliation(s)
- Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Vienna, Austria
| | - Enikö Hermann
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Vienna, Austria
| | - Angela L Rasmussen
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
2
|
Yamaji R, Zhang W, Kamata A, Adlhoch C, Swayne DE, Pereyaslov D, Wang D, Neumann G, Pavade G, Barr IG, Peiris M, Webby RJ, Fouchier RAM, Von Dobschütz S, Fabrizio T, Shu Y, Samaan M. Pandemic risk characterisation of zoonotic influenza A viruses using the Tool for Influenza Pandemic Risk Assessment (TIPRA). THE LANCET. MICROBE 2025; 6:100973. [PMID: 39396528 PMCID: PMC11876097 DOI: 10.1016/j.lanmic.2024.100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
A systematic risk assessment approach is essential for evaluating the relative risk of influenza A viruses (IAVs) with pandemic potential. To achieve this, the Tool for Influenza Pandemic Risk Assessment (TIPRA) was developed under the Global Influenza Programme of WHO. Since its release in 2016 and update in 2020, TIPRA has been used to assess the pandemic risk of 11 zoonotic IAVs across ten evaluation rounds. Notably, A(H7N9), A(H9N2), and A(H5) clade 2.3.4.4 viruses were re-evaluated owing to changes in epidemiological characteristics or virus properties. A(H7N9) viruses had the highest relative risk at the time of assessment, highlighting the importance of continuous monitoring and reassessment as changes in epidemiological trends within animal and human populations can alter risk profiles. The knowledge gaps identified throughout the ten risk assessments should help to guide the efficient use of resources for future research, including surveillance. The TIPRA tool reflects the One Health approach and has proven crucial for closely monitoring virus dynamics in both human and non-human populations to enhance preparedness for potential IAV pandemics.
Collapse
Affiliation(s)
- Reina Yamaji
- Global Influenza Programme, Epidemic and Pandemic Preparedness and Prevention, WHO Emergency Programme, World Health Organization, Geneva, Switzerland
| | - Wenqing Zhang
- Global Influenza Programme, Epidemic and Pandemic Preparedness and Prevention, WHO Emergency Programme, World Health Organization, Geneva, Switzerland
| | - Akiko Kamata
- The Food and Agriculture Organization of the UN (FAO), Rome, Italy
| | - Cornelia Adlhoch
- European Centre for Disease Prevention and Control, Solna, Sweden
| | | | - Dmitriy Pereyaslov
- Global Influenza Programme, Epidemic and Pandemic Preparedness and Prevention, WHO Emergency Programme, World Health Organization, Geneva, Switzerland
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, China CDC, Changping District, Beijing, China
| | - Gabriele Neumann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Sophie Von Dobschütz
- The Food and Agriculture Organization of the UN (FAO), Rome, Italy; Emerging Diseases and Zoonoses Unit, Department for Epidemic and Pandemic Preparedness and Prevention, World Health Organization, Geneva, Switzerland
| | - Thomas Fabrizio
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yuelong Shu
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Magdi Samaan
- Global Influenza Programme, Epidemic and Pandemic Preparedness and Prevention, WHO Emergency Programme, World Health Organization, Geneva, Switzerland.
| |
Collapse
|
3
|
Frey SE, Brady R, Jackson L, Goepfert P, El Sahly HM, Atmar RL, Rupp R, Creech CB, Abate G, Paulsen G, Weiss J, Wegel A, Roberts PC. Safety and immunogenicity of ascending doses of influenza A(H7N9) inactivated vaccine with or without MF59®. Vaccine 2025; 47:126702. [PMID: 39798434 DOI: 10.1016/j.vaccine.2024.126702] [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: 10/07/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
INTRODUCTION While it remains impossible to predict the timing of the next influenza pandemic, novel avian influenza A viruses continue to be considered a significant threat. METHODS A Phase II study was conducted in healthy adults aged 18-64 years to assess the safety and immunogenicity of two intramuscular doses of pre-pandemic 2017 influenza A(H7N9) inactivated vaccine administered 21 days apart. Participants were randomized (n = 105 in each of Arms 1-3) to receive 3.75 μg, 7.5 μg or 15 μg of hemagglutinin (HA) with MF59® adjuvant, or 15 μg of HA unadjuvanted vaccine (n = 57, Arm 4). RESULTS The three MF59 adjuvanted vaccines and the 15 μg unadjuvanted vaccine were safe and well-tolerated. Little antibody activity was detected against the A(H7N9) vaccine antigen after the first vaccination across study Arms. After second vaccination, the three adjuvanted Arms showed increases in hemagglutination inhibition (HAI), neutralizing (Neut), and neuraminidase inhibition (NAI) geometric mean titers (GMT), peaking at 21 days post second vaccination. The percentage of participants with titer ≥1:40 and seroconversion rates for HAI were 30-43 % and 0 for the adjuvanted Arms and the unadjuvanted Arm, respectively. Antibody responses against antigenically drifted A(H7N9) strains A/Shanghai/2/2013 and A/Guangdong/17SF003/2016 showed similar trends. Exploratory linear modeling of HAI and Neut responses post second vaccination revealed significantly lower log antibody titers among older participants (aged 35-49 and 50-64 years) compared to participants aged 18-34 years after adjusting for study vaccination, BMI, sex, and prior seasonal influenza vaccination. Post second vaccination, participants who received seasonal influenza vaccination in at least one of the two previous seasons had significantly lower log antibody titers than participants who did not. CONCLUSION Adjuvanted doses of vaccine provided higher antibody responses, on average, than the 15 μg unadjuvanted vaccine. Proportion of participants achieving seroconversion and antibody titers ≥40 remained below 50 % in all study Arm.
Collapse
MESH Headings
- Humans
- Influenza Vaccines/immunology
- Influenza Vaccines/adverse effects
- Influenza Vaccines/administration & dosage
- Adult
- Squalene/administration & dosage
- Squalene/adverse effects
- Squalene/immunology
- Polysorbates/administration & dosage
- Polysorbates/adverse effects
- Female
- Male
- Antibodies, Viral/blood
- Young Adult
- Influenza A Virus, H7N9 Subtype/immunology
- Middle Aged
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Adolescent
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/adverse effects
- Vaccines, Inactivated/administration & dosage
- Hemagglutination Inhibition Tests
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/adverse effects
- Immunogenicity, Vaccine
- Antibodies, Neutralizing/blood
- Adjuvants, Vaccine/administration & dosage
- Vaccination/methods
Collapse
Affiliation(s)
- Sharon E Frey
- Saint Louis University Center for Vaccine Development, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Rebecca Brady
- Cincinnati Children's Hospital Medical Center, Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lisa Jackson
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, USA
| | - Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hana M El Sahly
- Departments of Molecular Virology and Microbiology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Robert L Atmar
- Departments of Molecular Virology and Microbiology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Richard Rupp
- Sealy Institute for Vaccine Science, The University of Texas Medical Branch, Galveston, TX, USA
| | - C Buddy Creech
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Getahun Abate
- Saint Louis University Center for Vaccine Development, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Grant Paulsen
- Cincinnati Children's Hospital Medical Center, Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | | | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Sanz-Muñoz I, Sánchez-Martínez J, Rodríguez-Crespo C, Concha-Santos CS, Hernández M, Rojo-Rello S, Domínguez-Gil M, Mostafa A, Martinez-Sobrido L, Eiros JM, Nogales A. Are we serologically prepared against an avian influenza pandemic and could seasonal flu vaccines help us? mBio 2025; 16:e0372124. [PMID: 39745389 PMCID: PMC11796349 DOI: 10.1128/mbio.03721-24] [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: 12/09/2024] [Indexed: 02/06/2025] Open
Abstract
The current situation with H5N1 highly pathogenic avian influenza virus (HPAI) is causing a worldwide concern due to multiple outbreaks in wild birds, poultry, and mammals. Moreover, multiple zoonotic infections in humans have been reported. Importantly, HPAI H5N1 viruses with genetic markers of adaptation to mammals have been detected. Together with HPAI H5N1, avian influenza viruses H7N9 (high and low pathogenic) stand out due to their high mortality rates in humans. This raises the question of how prepared we are serologically and whether seasonal vaccines are capable of inducing protective immunity against these influenza subtypes. An observational study was conducted in which sera from people born between years 1925-1967, 1968-1977, and 1978-1997 were collected before or after 28 days or 6 months post-vaccination with an inactivated seasonal influenza vaccine. Then, hemagglutination inhibition, viral neutralization, and immunoassays were performed to assess the basal protective immunity of the population as well as the ability of seasonal influenza vaccines to induce protective responses. Our results indicate that subtype-specific serological protection against H5N1 and H7N9 in the representative Spanish population evaluated was limited or nonexistent. However, seasonal vaccination was able to increase the antibody titers to protective levels in a moderate percentage of people, probably due to cross-reactive responses. These findings demonstrate the importance of vaccination and suggest that seasonal influenza vaccines could be used as a first line of defense against an eventual pandemic caused by avian influenza viruses, to be followed immediately by the use of more specific pandemic vaccines.IMPORTANCEInfluenza A viruses (IAV) can infect and replicate in multiple mammalian and avian species. Avian influenza virus (AIV) is a highly contagious viral disease that occurs primarily in poultry and wild water birds. Due to the lack of population immunity in humans and ongoing evolution of AIV, there is a continuing risk that new IAV could emerge and rapidly spread worldwide, causing a pandemic, if the ability to transmit efficiently among humans was gained. The aim of this study is to analyze the basal protection and presence of antibodies against IAV H5N1 and H7N9 subtypes in the population from different ages. Moreover, we have evaluated the humoral response after immunization with a seasonal influenza vaccine. This study is strategically important to evaluate the level of population immunity that is a major factor when assessing the impact that an emerging IAV strain would have, and the role of seasonal vaccines to mitigate the effects of a pandemic.
Collapse
MESH Headings
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Influenza, Human/epidemiology
- Influenza, Human/virology
- Humans
- Antibodies, Viral/blood
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/genetics
- Animals
- Male
- Influenza in Birds/prevention & control
- Influenza in Birds/immunology
- Influenza in Birds/epidemiology
- Female
- Cross Reactions
- Middle Aged
- Aged
- Spain/epidemiology
- Pandemics/prevention & control
- Seasons
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccination
- Hemagglutination Inhibition Tests
- Adult
Collapse
Affiliation(s)
- Iván Sanz-Muñoz
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León, ICSCYL, Soria, Spain
- CIBERINFEC, Centro de Investigacion Biomedica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Sánchez-Martínez
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León, ICSCYL, Soria, Spain
| | - Carla Rodríguez-Crespo
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León, ICSCYL, Soria, Spain
| | | | - Marta Hernández
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Department of Microbiology, Universidad de Valladolid, Valladolid, Spain
| | - Silvia Rojo-Rello
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Microbiology Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Marta Domínguez-Gil
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Microbiology Unit, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - Ahmed Mostafa
- Texas Biomedical Research Institute, San Antonio, Texas, USA
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | | | - Jose M. Eiros
- National Influenza Centre, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
- Department of Microbiology, Universidad de Valladolid, Valladolid, Spain
- Microbiology Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
- Microbiology Unit, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, Madrid, Spain
- Center for Influenza Disease and Emergence Response (CIDER), Madrid, Spain
| |
Collapse
|
5
|
Zhu W, Dong C, Wei L, Kim JK, Wang BZ. Inverted HA-EV immunization elicits stalk-specific influenza immunity and cross-protection in mice. Mol Ther 2025; 33:485-498. [PMID: 39741410 PMCID: PMC11852689 DOI: 10.1016/j.ymthe.2024.12.052] [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: 08/20/2024] [Revised: 12/02/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Enhancing protective immunity in the respiratory tract is crucial to combat influenza infection and transmission. Developing mucosal universal influenza vaccines requires effective delivery platforms to overcome the respiratory mucosal barrier and stimulate appropriate innate immune reactions, thereby bridging adaptive immune responses with minimal necessary inflammation. Meanwhile, the vaccine platforms must be biocompatible. This study employed cell-derived extracellular vesicles (EVs) as a mucosal universal influenza vaccine platform. By conjugating influenza hemagglutinin (HA) onto EV surfaces through HA-receptor interaction, we achieved an upside-down (inverted) influenza HA configuration that exposed the conserved HA stalk region while partially hiding the globular head domain. Intranasal immunization with the resulting EVs induced robust HA stalk- and virus-specific serum antibody and mucosal immune responses in mice, protecting against heterologous virus infection. Notably, EVs derived from the lung epithelial cell line A549 induced superior cross-reactive antibodies and enhanced protection upon intranasal immunization. EVs conjugating multivalent HA elicited broadly cross-reactive antibody and cellular responses against different influenza strains. Our results demonstrated that EVs conjugating multiple inverted HAs represented an effective strategy for developing a mucosal universal influenza vaccine.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
6
|
Lin TH, Lee CCD, Fernández-Quintero ML, Ferguson JA, Han J, Zhu X, Yu W, Guthmiller JJ, Krammer F, Wilson PC, Ward AB, Wilson IA. Structurally convergent antibodies derived from different vaccine strategies target the influenza virus HA anchor epitope with a subset of V H3 and V K3 genes. Nat Commun 2025; 16:1268. [PMID: 39894881 PMCID: PMC11788443 DOI: 10.1038/s41467-025-56496-4] [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: 10/05/2024] [Accepted: 01/21/2025] [Indexed: 02/04/2025] Open
Abstract
H1N1 influenza viruses are responsible for both seasonal and pandemic influenza. The continual antigenic shift and drift of these viruses highlight the urgent need for a universal influenza vaccine to elicit broadly neutralizing antibodies (bnAbs). Identification and characterization of bnAbs elicited in natural infection and immunization to influenza virus hemagglutinin (HA) can provide insights for development of a universal influenza vaccine. Here, we structurally and biophysically characterize four antibodies that bind to a conserved region on the HA membrane-proximal region known as the anchor epitope. Despite some diversity in their VH and VK genes, the antibodies interact with the HA through germline-encoded residues in HCDR2 and LCDR3. Somatic mutations on HCDR3 also contribute hydrophobic interactions with the conserved HA epitope. This convergent binding mode provides extensive neutralization breadth against H1N1 viruses and suggests possible countermeasures against H1N1 viruses.
Collapse
MESH Headings
- Influenza Vaccines/immunology
- Epitopes/immunology
- Epitopes/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Humans
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Animals
Collapse
Affiliation(s)
- Ting-Hui Lin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Chang-Chun David Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Monica L Fernández-Quintero
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - James A Ferguson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Julianna Han
- 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
| | - Wenli Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jenna J Guthmiller
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Florian Krammer
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ignaz Semmelweis Institute, Medical University of Vienna, Vienna, Austria
| | - Patrick C Wilson
- Drukier Institute for Children's Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
7
|
Gu J, Yan Y, Zeng Z, Liu D, Hu J, Hu S, Wang X, Gu M, Liu X. Hemagglutinin with a polybasic cleavage site confers high virulence on H7N9 avian influenza viruses. Poult Sci 2025; 104:104832. [PMID: 39862488 PMCID: PMC11803838 DOI: 10.1016/j.psj.2025.104832] [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: 11/21/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
H7N9 avian influenza virus (AIV) first emerged in February 2013 in China, and early isolates were all low pathogenic (LP). After circulation for a few years in live poultry markets of China, LP H7N9 AIVs evolved into a highly pathogenic (HP) form in late 2016. Deduced amino acid sequence analysis of hemagglutinin (HA) gene revealed that all HP H7N9 AIVs have obtained four-amino-acid insertion at position 339-342 (H7 numbering), making the cleavage site from a monobasic motif (LP AIVs) to a polybasic form (HP AIVs). Notably, the polybasic cleavage site motifs are diversified, of which PEVPKRKRTAR↓GLF motif is prevalent. To elucidate the reasons accounting for its dominance, recombinant H7N9 virus carrying PEVPKRKRTAR↓GLF (rJT157-2) motif was generated based on LP H7N9 virus A/chicken/Eastern China/JT157/2016 (JT157). Besides, another two viruses containing PEVPKGKRTAR↓GLF (rJT157-1) and PEIPKRKRTAR↓GLF (rJT157-3) cleavage site motifs were also constructed as comparisons. We found that rJT157-2 showed better biological characterizations in vitro including replication kinetics, plaque size, thermal and acid stability. In addition, animal experiments demonstrated that rJT157-2 was more pathogenic to both chickens and mice with higher virus titers and induced more severe changes in the lungs. These results suggested that HP H7N9 viruses carrying PEVPKRKRTAR↓GLF motif in the HA cleavage site were most likely adaptive mutants during the evolution of H7N9 AIVs.
Collapse
Affiliation(s)
- Jinyuan Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; Shandong Engineering Research Center of Swine Health Data and Intelligent Monitoring, Dezhou University, Dezhou, PR China
| | - Yayao Yan
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Zixiong Zeng
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Dong Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, PR China; Jiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, PR China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, PR China; Jiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, PR China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, PR China; Jiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, PR China
| | - Min Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, PR China; Jiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, PR China.
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, PR China; Jiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, PR China.
| |
Collapse
|
8
|
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
|
9
|
Zhao B, Sun Z, Wang S, Shi Z, Jiang Y, Wang X, Deng G, Jiao P, Chen H, Wang J. Structural basis of different neutralization capabilities of monoclonal antibodies against H7N9 virus. J Virol 2025; 99:e0140024. [PMID: 39704525 PMCID: PMC11784312 DOI: 10.1128/jvi.01400-24] [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: 08/24/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
Neutralizing antibodies (nAbs) are important for the treatment of emerging viral diseases and for effective vaccine development. In this study, we generated and evaluated three nAbs (1H9, 2D7, and C4H4) against H7N9 influenza viruses and found that they differ in their ability to inhibit viral attachment, membrane fusion, and egress. We resolved the cryo-electron microscopy (cryo-EM) structures of H7N9 hemagglutinin (HA) alone and in complex with the nAb antigen-binding fragments (Fabs) and identified the HA head-located epitope for each nAb, thereby revealing the molecular basis and key residues that determine the differences in these nAbs in neutralizing H7N9 viruses. Moreover, we found that the humanized nAb CC4H4 provided complete protection in mice against death caused by a lethal H7N9 virus infection, even when nAb was given 3 days after the mice were infected. These findings provide new insights into the neutralizing mechanism and structural basis for the rational design of H7N9 virus vaccines and therapeutics.IMPORTANCEH7N9 viruses have caused severe infections in both birds and humans since their emergence in early 2013 in China. Their persistent presence and variation in avian populations pose a significant threat to both poultry and humans. There are no treatments for human infections. In this study, we thoroughly investigated the neutralization mechanisms, structural basis, and therapeutic effects of three nAbs (1H9, 2D7, and C4H4) against H7N9 viruses. We revealed the molecular determinants underlying the varied performances of the three nAbs in neutralizing H7N9 viruses and protecting H7N9-infected mice. These insights provide a solid foundation for the rational design of vaccines and therapeutics against H7N9 viruses.
Collapse
MESH Headings
- Influenza A Virus, H7N9 Subtype/immunology
- Animals
- Antibodies, Neutralizing/immunology
- Mice
- Antibodies, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/chemistry
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Humans
- Cryoelectron Microscopy
- Epitopes/immunology
- Epitopes/chemistry
- Mice, Inbred BALB C
- Influenza, Human/immunology
- Influenza, Human/virology
- Influenza, Human/prevention & control
- Female
- Influenza Vaccines/immunology
Collapse
Affiliation(s)
- Bingbing Zhao
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zhenzhao Sun
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Shida Wang
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zhibin Shi
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, 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 & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xiurong Wang
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, 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 & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Peirong Jiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jingfei Wang
- State Key Laboratory for Animal Disease Control and Prevention & National Data Center for Animal Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| |
Collapse
|
10
|
Boyd DF, Jordan SV, Balachandran S. ZBP1-driven cell death in severe influenza. Trends Microbiol 2025:S0966-842X(24)00324-X. [PMID: 39809680 DOI: 10.1016/j.tim.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Influenza A virus (IAV) infections can cause life-threatening illness in humans. The severity of disease is directly linked to virus replication in the alveoli of the lower respiratory tract. In particular, the lytic death of infected alveolar epithelial cells (AECs) is a major driver of influenza severity. Recent studies have begun to define the molecular mechanisms by which IAV triggers lytic cell death. Z-form nucleic-acid-binding protein 1 (ZBP1) senses replicating IAV and drives programmed cell death (PCD) in infected cells, including apoptosis and necroptosis in AECs and pyroptosis in myeloid cells. Necroptosis and pyroptosis, both lytic forms of death, contribute to pathogenesis during severe infections. Pharmacological blockade of necroptosis shows strong therapeutic potential in mouse models of lethal influenza. We suggest that targeting ZBP1-initiated necroinflammatory cell lysis, either alone or in combination antiviral drugs, will provide clinical benefit in severe influenza.
Collapse
Affiliation(s)
- David F Boyd
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA.
| | - Summer Vaughn Jordan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA
| | | |
Collapse
|
11
|
Jallow MM, Diagne MM, Ndione MHD, Barry MA, Ndiaye NK, Kiori DE, Mendy MP, Goudiaby D, Fall G, Fall M, Dia N. Genetic and Molecular Characterization of Avian Influenza A(H9N2) Viruses from Live Bird Markets (LBM) in Senegal. Viruses 2025; 17:73. [PMID: 39861862 PMCID: PMC11769557 DOI: 10.3390/v17010073] [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: 11/20/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Despite extensive experience with influenza surveillance in humans in Senegal, there is limited knowledge about the actual situation and genetic diversity of avian influenza viruses (AIVs) circulating in the country, hindering control measures and pandemic risk assessment. Therefore, as part of the "One Health" approach to influenza surveillance, we conducted active AIV surveillance in two live bird markets (LBMs) in Dakar to better understand the dynamics and diversity of influenza viruses in Senegal, obtain genetic profiles of circulating AIVs, and assess the risk of emergence of novel strains and their transmission to humans. Cloacal swabs from poultry and environmental samples collected weekly from the two LBMs were screened by RT-qPCR for H5, H7, and H9 AIVs. Subsequently, a subset of H9-positive samples was selected for whole sequencing. From December 2023 to October 2024, 499 samples were tested, and AIV was detected in 58.3% of them. Among these, A/H9N2 was the only subtype detected in both markets, with a detection rate of 47.7% (82/172) in Thiaroye and 35.3% (42/119) in Tilene, resulting in an overall positivity rate of 42.6% (124/291). Genome sequencing of 22 A/H9N2 isolates, including 11 poultry drinking water samples, 7 carcass wash water samples, 3 fecal samples, and 1 cloacal swab, yielded 7 complete and 15 partial genomic sequences. Phylogenetic analyses of the resulting sequences showed that the A/H9N2 isolates obtained in this study formed a monophyletic cluster and were closely related to the Senegalese human strain (A/Senegal/0243/2019) identified through the national influenza sentinel surveillance program. These strains were also closely related to the A/H9N2 viruses of the G1 lineage circulating in neighboring countries, suggesting cross-border transmission. The A/H9N2 strains carried the low pathogenicity RSSR/GLF motif at the HA cleavage site and possessed several key amino acid mutations, including HA-I155T and HA-Q226L, which are associated with human host adaptation, PB2-T105V, PB2-A661T, and PB2-A588V, which are linked to the human-to-human transmission and increased polymerase activity, NS2-T14M, NS2-M100I, NS1-I106M, NS1-V222M, NS1-E223A, NS1-I226V, NS1-E227G, and NS1-P228S, which are known to alter virulence (increased or reduced) in humans or mice, and M2-S31N, which promotes drug resistance. Seven potential N-glycosylation sites were predicted in the HA protein and six in the NA protein. The selection pressure analysis revealed that the A/H9N2 isolates were primarily under neutral evolution or purifying selection pressure. Overall, our findings highlight the potential for cross-species transmission of Senegalese A/H9N2 viruses, emphasizing the need for sustained monitoring of these viruses in both animal and human populations.
Collapse
Affiliation(s)
- Mamadou Malado Jallow
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
- Département de Biologie Animale, Faculté des Sciences et Techniques, Université Cheikh Anta DIOP de Dakar, Dakar BP 206, Senegal;
| | - Moussa Moise Diagne
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Marie Henriette Dior Ndione
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Mamadou Aliou Barry
- Unité d’Epidémiologie des Maladies Infectieuses, Institut Pasteur de Dakar, Dakar BP 220, Senegal;
| | - Ndiendé Koba Ndiaye
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Davy Evrard Kiori
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Marie Pedapa Mendy
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Déborah Goudiaby
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Gamou Fall
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| | - Malick Fall
- Département de Biologie Animale, Faculté des Sciences et Techniques, Université Cheikh Anta DIOP de Dakar, Dakar BP 206, Senegal;
| | - Ndongo Dia
- Département de Virologie, Institut Pasteur de Dakar, Dakar BP 220, Senegal; (M.M.J.); (M.M.D.); (M.H.D.N.) ; (N.K.N.); (D.E.K.); (M.P.M.); (D.G.) ; (G.F.)
| |
Collapse
|
12
|
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
|
13
|
Zhang X, Skarlupka AL, Shi H, Ross TM. COBRA N2 NA vaccines induce protective immune responses against influenza viral infection. Hum Vaccin Immunother 2024; 20:2403175. [PMID: 39291424 DOI: 10.1080/21645515.2024.2403175] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024] Open
Abstract
Influenza neuraminidase (NA) is a promising target for a broadly protective vaccine. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to develop N2 NA vaccine candidates. The unique wild type (WT) N2 sequences of human and swine influenza strains isolated between 1957 and 2019 were used to design the COBRA N2-A NA vaccine, while the unique WT N2 sequences of human influenza strains isolated between 2000 and 2019 were used to design the COBRA N2-B NA vaccine. Sera collected from COBRA N2 NA vaccinated mice showed more broadly reactive antibody responses against a broad panel of H×N2 influenza virus strains than sera collected from mice vaccinated with WT N2 NA vaccines. Antibodies elicited by COBRA or WT N2 NA antigens cross react with recent human H3N2 influenza viruses from different clades, while the antibodies elicited by A/Switzerland/9715293/2013 hemagglutinin (HA) reacted with viruses from the same clade. Furthermore, mice vaccinated with COBRA N2-B NA vaccine had lower viral lung titers compared to mock vaccinated mice when challenged with human H3N2 influenza viruses. Thus, the COBRA N2 NA vaccines elicit broadly protective murine anti-NA antibodies against multiple strains across subtypes and the viral loads were significantly decreased in the lungs of the mice in the COBRA N2 NA vaccine groups, compared to the mice in the mock vaccinated group, indicating that the COBRA-based N2 subtype NA vaccines have a potential to be a component in a universal influenza vaccine.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Amanda L Skarlupka
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
14
|
Escuyer KL, Gowie DL, St. George K. Influenza Virus Surveillance from the 1918 Influenza Pandemic to the 2020 Coronavirus Pandemic in New York State, USA. Viruses 2024; 16:1952. [PMID: 39772258 PMCID: PMC11680092 DOI: 10.3390/v16121952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
A historical perspective of more than one hundred years of influenza surveillance in New York State demonstrates the progression from anecdotes and case counts to next-generation sequencing and electronic database management, greatly improving pandemic preparedness and response. Here, we determined if influenza virologic surveillance at the New York State public health laboratory (NYS PHL) tests sufficient specimen numbers within preferred confidence limits to assess situational awareness and detect novel viruses that pose a pandemic risk. To this end, we analyzed retrospective electronic data on laboratory test results for the influenza seasons 1997-1998 to 2021-2022 according to sample sizes recommended in the Influenza Virologic Surveillance Right Size Roadmap issued by the Association of Public Health Laboratories and Centers for Disease Control and Prevention. Although data solely from specimens submitted to the NYS PHL were insufficient to meet surveillance goals, when supplemented with testing data from clinical laboratories participating in surveillance programs, the recommended surveillance goals were achieved. Despite the sudden decline in influenza cases in 2020-2021, impacted by the COVID-19 mitigation measures, the dramatic increases in influenza cases surrounding the coronavirus pandemic reveal that influenza remains a national and international public health threat. Sample submissions to public health laboratories must be encouraged to facilitate monitoring for emerging viruses and preparedness for another pandemic.
Collapse
Affiliation(s)
- Kay L. Escuyer
- Wadsworth Center, David Axelrod Institute, New York State Department of Health, Albany, NY 12208, USA;
| | - Donna L. Gowie
- Bureau of Communicable Disease Control, Empire State Plaza, New York State Department of Health, Albany, NY 12237, USA;
| | - Kirsten St. George
- Wadsworth Center, David Axelrod Institute, New York State Department of Health, Albany, NY 12208, USA;
| |
Collapse
|
15
|
Zhou Y, Li Y, Chen H, Shu S, Li Z, Sun H, Sun Y, Liu J, Lu L, Pu J. Origin, spread, and interspecies transmission of a dominant genotype of BJ/94 lineage H9N2 avian influenza viruses with increased threat. Virus Evol 2024; 10:veae106. [PMID: 39735714 PMCID: PMC11673197 DOI: 10.1093/ve/veae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/31/2024] Open
Abstract
The H9N2 subtype of avian influenza viruses (AIVs) is widely prevalent in poultry and wild birds globally, with occasional transmission to humans. In comparison to other H9N2 lineages, the BJ/94 lineage has raised more public health concerns; however, its evolutionary dynamics and transmission patterns remain poorly understood. In this study, we demonstrate that over three decades (1994-2023), BJ/94 lineage has undergone substantial expansion in its geographical distribution, interspecies transmission, and viral reassortment with other AIV subtypes, increasing associated public health risks. These changes were primarily driven by the emergence of a dominant genotype G57. In the first decade, G57 emerged in East China and rapidly adapted to chickens and spread across China. Since 2013, the G57 genotype has expanded beyond China into eight other countries and reassorted with various AIV subtypes to form new zoonotic reassortants. Chickens have played a key role in the generation and circulation of the G57 viruses, with ducks and other poultry species likely assuming an increasingly importantly role. Over the past decade, G57 has been more frequently detected in wild birds, mammals, and humans. Additionally, Vietnam has emerged as a new hotspot for the international spread of G57. Our results suggest that the BJ/94 lineage H9N2 virus may continue to overcome geographical and species barriers, with potentially more severe consequences.
Collapse
Affiliation(s)
- Yong Zhou
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Yudong Li
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Hongzhuang Chen
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Sicheng Shu
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Zhixin Li
- Ningxia Hui Autonomous Region Animal Disease Prevention and Control Center, No. 411, Mancheng South Street, Jinfeng District, Yinchuan City, Ningxia Hui Autonomous Region, Yinchuan 750011, China
| | - Honglei Sun
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Yipeng Sun
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Jinhua Liu
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Lu Lu
- Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Edinburgh EH2 59RG, United Kingdom
| | - Juan Pu
- National Key Laboratory of Veterinary Public Health and Safety, 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, 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| |
Collapse
|
16
|
He F, Yu H, Liu L, Li X, Xing Y, Yang L, Yang P, Zhu L, Li Z. Antigenicity and genetic properties of an Eurasian avian-like H1N1 swine influenza virus in Jiangsu Province, China. BIOSAFETY AND HEALTH 2024; 6:319-326. [PMID: 40078985 PMCID: PMC11894957 DOI: 10.1016/j.bsheal.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 03/14/2025] Open
Abstract
Pigs are vital genetic mixing vessels for human and avian influenza viruses because their tracheal epitheliums possess both sialic acid α-2,6-Gal and α-2,3-Gal receptors. Cross-species transmission of influenza A viruses from swine to humans occurs occasionally. The first case of human infection with the Eurasian avian-like H1N1 swine influenza virus (EAH1N1 SIVs) genotype G4 was detected in Jiangsu Province, China, in February 2023, and backtracking epidemiological investigations did not reveal a clear source of the infection. The hemagglutination (HA) and neuraminidase (NA) amino acid variant sites, antiviral drug susceptibility, and antigenic variation of the isolated A/Jiangsu/27271/2023 (JS/27271/23) virus were analyzed, and we evaluated the protective effect of sera collected from occupationally exposed populations in 2024 against the virus. Compared with the vaccine strain, the nucleotide sequence similarities of JS/27271/23 HA and NA were 96.5 % and 95.2 %, respectively. JS/27271/23 was sensitive to polymerase inhibitors (favipiravir and baloxavir), and the antigenicity of its HA protein was 8-fold different from that of the vaccine strain. The percentage of occupationally exposed population with antibody titers of ≥ 40 against A/Hunan/42443/2015 (HN/42443/15) and A/Jiangsu/1/2011 (JS/1/11) were 7.25 % and 2.25 %, respectively, and the geometric mean titers (GMT) were 6.24 and 5.34, respectively. Out of 400 serum samples examined, none had antibody titers of ≥ 40 against JS/27271/23. This suggests that low serum levels of antibodies to EAH1N1 SIVs in occupationally exposed populations may not provide adequate protection because of significant differences in amino acid sites and antigenicity between this virus and the current vaccine strain of EAH1N1 SIVs. There is no evidence of human-to-human transmission of EAH1N1 SIVs. Therefore, surveillance for EAH1N1 SIVs and the development of new vaccine strains are required.
Collapse
Affiliation(s)
- Fang He
- Huai’an Center for Disease Control and Prevention, Huai’an 223299, China
| | - Huiyan Yu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210008, China
| | - Liqi Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiyan Li
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yadong Xing
- Huai’an Center for Disease Control and Prevention, Huai’an 223299, China
| | - Lei Yang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Pengfei Yang
- Huai’an Center for Disease Control and Prevention, Huai’an 223299, China
| | - Liguo Zhu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210008, China
- Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210008, China
| | - Zi Li
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
17
|
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
|
18
|
Tang J, Zou SM, Zhou JF, Gao RB, Xin L, Zeng XX, Huang WJ, Li XY, Cheng YH, Liu LQ, Xiao N, Wang DY. R229I substitution from oseltamivir induction in HA1 region significantly increased the fitness of a H7N9 virus bearing NA 292K. Emerg Microbes Infect 2024; 13:2373314. [PMID: 38922326 PMCID: PMC467099 DOI: 10.1080/22221751.2024.2373314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
The proportion of human isolates with reduced neuraminidase inhibitors (NAIs) susceptibility in highly pathogenic avian influenza (HPAI) H7N9 virus was high. These drug-resistant strains showed good replication capacity without serious loss of fitness. In the presence of oseltamivir, R229I substitution were found in HA1 region of the HPAI H7N9 virus before NA R292K appeared. HPAI H7N9 or H7N9/PR8 recombinant viruses were developed to study whether HA R229I could increase the fitness of the H7N9 virus bearing NA 292K. Replication efficiency was assessed in MDCK or A549 cells. Neuraminidase enzyme activity and receptor-binding ability were analyzed. Pathogenicity in C57 mice was evaluated. Antigenicity analysis was conducted through a two-way HI test, in which the antiserum was obtained from immunized ferrets. Transcriptomic analysis of MDCK infected with HPAI H7N9 24hpi was done. It turned out that HA R229I substitution from oseltamivir induction in HA1 region increased (1) replication ability in MDCK(P < 0.05) and A549(P < 0.05), (2) neuraminidase enzyme activity, (3) binding ability to both α2,3 and α2,6 receptor, (4) pathogenicity to mice(more weight loss; shorter mean survival day; viral titer in respiratory tract, P < 0.05; Pathological changes in pneumonia), (5) transcriptome response of MDCK, of the H7N9 virus bearing NA 292K. Besides, HA R229I substitution changed the antigenicity of H7N9/PR8 virus (>4-fold difference of HI titre). It indicated that through the fine-tuning of HA-NA balance, R229I increased the fitness and changed the antigenicity of H7N9 virus bearing NA 292K. Public health attention to this mechanism needs to be drawn.
Collapse
MESH Headings
- Animals
- Oseltamivir/pharmacology
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/drug effects
- Influenza A Virus, H7N9 Subtype/pathogenicity
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/physiology
- Neuraminidase/genetics
- Neuraminidase/metabolism
- Dogs
- Virus Replication/drug effects
- Antiviral Agents/pharmacology
- Humans
- Mice
- Orthomyxoviridae Infections/virology
- Madin Darby Canine Kidney Cells
- A549 Cells
- Mice, Inbred C57BL
- Drug Resistance, Viral/genetics
- Amino Acid Substitution
- Influenza, Human/virology
- Ferrets
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Female
- Viral Proteins/genetics
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Jing Tang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Shu-Mei Zou
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Jian-Fang Zhou
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Rong-Bao Gao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Li Xin
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Xiao-Xu Zeng
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Wei-Juan Huang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Xi-Yan Li
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Yan-Hui Cheng
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Li-Qi Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Ning Xiao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| | - Da-Yan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Reference and Research on Influenza; Key Laboratory for Medical Virology and Viral Diseases, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing, People’s Republic of China
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Qu R, Chen M, Chen C, Cao K, Wu X, Zhou W, Qi J, Miao J, Yan D, Yang S. Risk distribution of human infections with avian influenza A (H5N1, H5N6, H9N2 and H7N9) viruses in China. Front Public Health 2024; 12:1448974. [PMID: 39512713 PMCID: PMC11540643 DOI: 10.3389/fpubh.2024.1448974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
Background This study aimed to investigate epidemiologic characteristics of major human infection with avian influenza and explore the factors underlying the spatial distributions, particularly H5N6 and H9N2, as H9N2 could directly infect mankind and contribute partial or even whole internal genes to generate novel human-lethal reassortants such as H5N6. They pose potential threats to public health and agriculture. Methods This study collected cases of H5N1, H5N6, H9N2, and H7N9 in China, along with data on ecoclimatic, environmental, social and demographic factors at the provincial level. Boosted regression tree (BRT) models, a popular approach to ecological studies, has been commonly used for risk mapping of infectious diseases, therefore, it was used to investigate the association between these variables and the occurrence of human cases for each subtype, as well as to map the probabilities of human infections. Results A total of 1,123 H5N1, H5N6, H9N2, and H7N9 human cases have been collected in China from 2011 to 2024. Factors including density of pig and density of human population emerged as common significant predictors for H5N1 (relative contributions: 5.3, 5.8%), H5N6 (10.8, 6.4%), H9N2 (11.2, 7.3%), and H7N9 (9.4, 8.0%) infection. Overall, each virus has its own ecological and social drivers. The predicted distribution probabilities for H5N1, H5N6, H9N2, and H7N9 presence are highest in Guangxi, Sichuan, Guangdong, and Jiangsu, respectively, with values of 0.86, 0.96, 0.93 and 0.99. Conclusion This study highlighted the important role of social and demographic factors in the infection of different avian influenza, and suggested that monitoring and control of predicted high-risk areas should be prioritized.
Collapse
Affiliation(s)
- Rongrong Qu
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengsha Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Can Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Kexin Cao
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyue Wu
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenkai Zhou
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxing Qi
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiani Miao
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dong Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shigui Yang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Han M, Lu Q, Wang D, Zhou K, Jia C, Teng L, Hamuti A, Peng X, Hu Y, Li W, Yue M, Li Y. Oral co-administration of Lactiplantibacillus plantarum 16 and Lacticaseibacillus rhamnosus P118 improves host defense against influenza A virus infection. J Virol 2024; 98:e0095024. [PMID: 39258911 PMCID: PMC11494971 DOI: 10.1128/jvi.00950-24] [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: 06/02/2024] [Accepted: 08/25/2024] [Indexed: 09/12/2024] Open
Abstract
Influenza is an important zoonotic disease that persistently threatens global public health. While it is widely acknowledged that probiotics can modulate the host response to protect the host against infectious disease, the prophylactic efficacy on respiratory viral infection and the detailed mechanism remains elusive. Lactobacillus, the most commonly used probiotic widely applied in food production, has garnered significant attention. In our study utilizing both C57BL/6 and BALB/c mouse models, we explored the protective effect against two strains of influenza virus, A/Mink/China/01/2014(H9N2) and A/California/04/2009(H1N1), through the administration of Lactiplantibacillus plantarum strain 16 (L. plantarum 16) and Lacticaseibacillus rhamnosus strain P118 (L. rhamnosus P118), aiming to identify robust probiotic strains with antiviral properties. Our findings indicate that administering L. plantarum 16 or L. rhamnosus P118 alone does not provide sufficient protection against influenza. However, the co-administration of L. plantarum 16 and L. rhamnosus P118 dramatically reduces viral titers in the respiratory tract and lung, thereby markedly alleviating the clinical symptoms, improving prognosis, and reducing mortality. The mechanisms underlying this effect involve the modulation of host gut microbiota and metabolism through the co-administration of L. plantarum 16 and L. rhamnosus P118, resulting in enrichment of Firmicutes and enhancement of phenylalanine-related metabolism, ultimately leading to an augmentation of the antiviral immune response. Notably, we identified that the circulating metabolic molecule 2-Hydroxycinnamic acid plays a significant role in combating influenza. Our data suggest the potential utility of L. plantarum 16 and L. rhamnosus P118 two-bacterium or 2-Hydroxycinnamic acid in preventing influenza.IMPORTANCEVaccination represents the most optimal strategy to control influenza. Nevertheless, influenza viruses constantly evolve due to antigenic drift and shift, leading to the need for regular updates on influenza vaccines. Additionally, vaccination failure poses significant challenges to influenza prevention. Therefore, it is essential and beneficial to identify novel or universal antiviral measures to protect against influenza. While cumulative data suggest that probiotics offer protection against infectious diseases, the specific mechanisms, such as the effective metabolites or components, remain largely unknown. Our research discovered the capacity of combinational two-bacterium Lactiplantibacillus plantarum 16 and Lacticaseibacillus rhamnosus P118 to fight against influenza infection in a mouse model. The protection may occur through modulating the host's gut microbiota and metabolism, further influencing the host's antiviral immune response. Notably, we have identified a novel metabolic molecule, 2-Hydroxycinnamic acid, capable of enhancing antiviral response and restricting viral replication in vivo.
Collapse
Affiliation(s)
- Meiqing Han
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Sanya, Hainan, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Qi Lu
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Sanya, Hainan, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Di Wang
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Kun Zhou
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Chenghao Jia
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Sanya, Hainan, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Lin Teng
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Azeguli Hamuti
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Xianqi Peng
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Yixiang Hu
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Sanya, Hainan, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
| | - Min Yue
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Sanya, Hainan, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yan Li
- Department of Veterinary Medicine and Institute of Preventive Veterinary Sciences, Zhejiang University College of Animal Sciences, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Sanya, Hainan, China
- MOA Key Laboratory of Animal Virology, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Odin M, Schmeisser F, Soto J, Weir J. A Rapid Virus-Free Method for Producing Influenza HA Immunogen Needed for Preparation of Influenza Vaccine Potency Antisera Reagents. Influenza Other Respir Viruses 2024; 18:e70024. [PMID: 39440693 PMCID: PMC11497102 DOI: 10.1111/irv.70024] [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: 08/02/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The potency of inactivated and recombinant influenza vaccines is measured using the single-radial immunodiffusion (SRID) assay. The strain-specific antigen and antibody potency reagents required for the assay are prepared and distributed by regulatory agencies to ensure vaccine standardization, but timely reagent production is always challenging. This poses unique concerns for rapid pandemic responses. Alternative methods have been described for generating strain-specific potency antibody reagents without the need for live influenza virus, but such methods are infrequently used, suggesting the need for additional antigen expression approaches. METHODS We describe a rapid process using a mammalian expression system to produce recombinant influenza hemagglutinin (rHA). This platform was used to generate rHA from two H5 clade 2.3.4.4 influenza viruses, in both soluble ectodomain or full-length HA forms, and a soluble ectodomain rHA from an influenza H2 virus. RESULTS The purified rHAs were used as immunogens to produce HA antibody reagents that were tested for suitability in the SRID assay to accurately measure the potency of inactivated pandemic influenza vaccines. Antibody reagents generated to either ectodomain or full-length rHA worked well in the SRID assay and resulted in vaccine potency values equivalent to those generated with standard reference antibodies. CONCLUSIONS The results demonstrate that rHA produced from a simple mammalian cell transfection method can be used to generate HA antibody suitable for use in the influenza vaccine SRID potency assay and suggest a practical means by which an extensive library of pandemic reagents can easily be prepared in advance of and during an influenza emergency.
Collapse
Affiliation(s)
- Marcus Odin
- Division of Viral Products, Laboratory of DNA Viruses, Center for Biologics Evaluation and ResearchFood and Drug AdministrationSilver SpringMarylandUSA
| | - Falko Schmeisser
- Division of Viral Products, Laboratory of DNA Viruses, Center for Biologics Evaluation and ResearchFood and Drug AdministrationSilver SpringMarylandUSA
| | - Jackeline Soto
- Division of Viral Products, Laboratory of DNA Viruses, Center for Biologics Evaluation and ResearchFood and Drug AdministrationSilver SpringMarylandUSA
| | - Jerry P. Weir
- Division of Viral Products, Laboratory of DNA Viruses, Center for Biologics Evaluation and ResearchFood and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
24
|
Daidoji T, Sadakane H, Garan K, Kawashita N, Arai Y, Watanabe Y, Nakaya T. The host tropism of current zoonotic H7N9 viruses depends mainly on an acid-labile hemagglutinin with a single amino acid mutation in the stalk region. PLoS Pathog 2024; 20:e1012427. [PMID: 39436936 PMCID: PMC11495601 DOI: 10.1371/journal.ppat.1012427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 07/15/2024] [Indexed: 10/25/2024] Open
Abstract
The incidence of human infection by zoonotic avian influenza viruses, especially H5N1 and H7N9 viruses, has increased. Current zoonotic H7N9 avian influenza viruses (identified since 2013) emerged during reassortment of viruses belonging to different subtypes. Despite analyses of their genetic background, we do not know why current H7N9 viruses are zoonotic. Therefore, there is a need to identify the factor(s) responsible for the extended host tropism that enables these viruses to infect humans as well as birds. To identify H7N9-specific amino acids that confer zoonotic properties on H7N9 viruses, we performed multiple alignment of the hemagglutinin (HA) amino acid sequences of A/Shanghai/1/2013 (H7N9) and A/duck/Zhejiang/12/2011(H7N3) (a putative, non- or less zoonotic HA donor to the zoonotic H7N9 virus). We also analyze the function of an H7N9 HA-specific amino acid with respect to HA acid stability, and evaluated the effect of acid stability on viral infectivity and virulence in a mouse model. HA2-116D, preserved in current zoonotic H7N9 viruses, was crucial for loss of HA acid stability. The acid-labile HA protein in H7 viruses played an important role in infection of human airway epithelial cells; HA2-116D contributed to infection and replication of H7 viruses. Finally, HA2-116D served as a H7 virulence factor in mice. These results suggest that acid-labile HA harboring HA2-116D confers zoonotic characteristics on H7N9 virus and that future novel zoonotic avian viruses could emerge from non-zoonotic H7 viruses via acquisition of mutations that remove HA acid stability.
Collapse
Affiliation(s)
- Tomo Daidoji
- Department of Pathobiology, School of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Sadakane
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kotaro Garan
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Yasuha Arai
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yohei Watanabe
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takaaki Nakaya
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
25
|
Sharma A, Chandrashekar CR, Krishna S, Sowdhamini R. Computational Analysis of the Accumulation of Mutations in Therapeutically Important RNA Viral Proteins During Pandemics with Special Emphasis on SARS-CoV-2. J Mol Biol 2024; 436:168716. [PMID: 39047897 DOI: 10.1016/j.jmb.2024.168716] [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: 04/07/2024] [Revised: 07/06/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Single stranded RNA viruses are primary causative agents for pandemics, causing extensive morbidity and mortality worldwide. A pivotal question in pandemic preparedness and therapeutic intervention is what are the specific mutations which are more likely to emerge during such global health crises? This study aims to identify markers for mutations with the highest probability of emergence in these pandemics, focusing on the SARS-CoV-2 spike protein, an essential and therapeutically significant viral protein, starting from sequence information from the onset of the pandemic until July 2022. Quite consistently, we observed that emerged mutations tended to demonstrate a high genetic score, which reflects high similarity of the type of codon required for translation between an amino acid and to the mutated one. Further, this pattern is also observed in therapeutically significant proteins of other ssRNA pandemic viruses, including influenza (HA, NA), spike proteins of Ebola, envelope of Dengue and Chikungunya. We propose that the genetic score serves as an initial indicator, preceding the actual impact of the mutation on viral fitness. Finally, we developed a comprehensive computational pipeline to further explore and predict the subsequent effects of mutations on viral fitness. We believe that our pipeline can narrow down and predict future mutations in therapeutically important viral proteins during a pandemic.
Collapse
Affiliation(s)
- Abhishek Sharma
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - C R Chandrashekar
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - Sudhir Krishna
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - Ramanathan Sowdhamini
- Molecular Biophysics Unit, Indian Institute of Science, Banagalore 560012, India; Institute of Bioinformatics and Applied Biotechnology, Bangalore, 560100, India.
| |
Collapse
|
26
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
27
|
Chen SS, Yang YL, Wang HY, Guo TK, Azeem RM, Shi CW, Yang GL, Huang HB, Jiang YL, Wang JZ, Cao X, Wang N, Zeng Y, Yang WT, Wang CF. CRISPR/Cas13a-based genome editing for establishing the detection method of H9N2 subtype avian influenza virus. Poult Sci 2024; 103:104068. [PMID: 39096825 PMCID: PMC11345561 DOI: 10.1016/j.psj.2024.104068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 08/05/2024] Open
Abstract
Avian influenza virus (AIV) subtype H9N2 has significantly threatened the poultry business in recent years by having become the predominant subtype in flocks of chickens, ducks, and pigeons. In addition, the public health aspects of H9N2 AIV pose a significant threat to humans. Early and rapid diagnosis of H9N2 AIV is therefore of great importance. In this study, a new method for the detection of H9N2 AIV based on fluorescence intensity was successfully established using CRISPR/Cas13a technology. The Cas13a protein was first expressed in a prokaryotic system and purified using nickel ion affinity chromatography, resulting in a high-purity Cas13a protein. The best RPA (recombinase polymerase amplification) primer pairs and crRNA were designed and screened, successfully constructing the detection of H9N2 AIV based on CRISPR/Cas13a technology. Optimal concentration of Cas13a and crRNA was determined to optimize the constructed assay. The sensitivity of the optimized detection system is excellent, with a minimum detection limit of 10° copies/μL and didn't react with other avian susceptible viruses, with excellent specificity. The detection method provides the basis for the field detection of the H9N2 AIV.
Collapse
Affiliation(s)
- Sha-Sha Chen
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yong-Lei Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Hong-Yun Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Tian-Kui Guo
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Riaz-M Azeem
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
28
|
Zhang Z, Lei Z. The Alarming Situation of Highly Pathogenic Avian Influenza Viruses in 2019-2023. Glob Med Genet 2024; 11:200-213. [PMID: 38947761 PMCID: PMC11213626 DOI: 10.1055/s-0044-1788039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Avian influenza viruses (AIVs) have the potential to cause severe illness in wild birds, domestic poultry, and humans. The ongoing circulation of highly pathogenic avian influenza viruses (HPAIVs) has presented significant challenges to global poultry industry and public health in recent years. This study aimed to elucidate the circulation of HPAIVs during 2019 to 2023. Specifically, we assess the alarming global spread and continuous evolution of HPAIVs. Moreover, we discuss their transmission and prevention strategies to provide valuable references for future prevention and control measures against AIVs.
Collapse
Affiliation(s)
- Zhiwei Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian Province, People's Republic of China
- Department of Industrial & Systems Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Zhao Lei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian Province, People's Republic of China
| |
Collapse
|
29
|
Chen X, Mu W, Shao Y, Peng L, Zhang R, Luo S, He X, Zhang L, He F, Li L, Wang R, Yang L, Xiang B. Genetic and molecular characterization of H9N2 avian influenza viruses in Yunnan Province, Southwestern China. Poult Sci 2024; 103:104040. [PMID: 39043028 PMCID: PMC11318558 DOI: 10.1016/j.psj.2024.104040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/02/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
The H9N2 subtype of the avian influenza virus (AIV) is widely prevalent in birds, threatening the poultry industry and providing genetic material for emerging human pathogens. The prevalence and genetic characteristics of H9N2 in Yunnan Province, China, are largely unknown. Samples were collected from live poultry markets (LPMs) and breeding farms in Yunnan Province. H9N2-positive samples were identified by polymerase chain reaction (PCR), with a high positivity rate of 42.86% in tissue samples. The positivity rate of swab samples in the LPMs in Kunming was 3.97% (17/564), but no AIV was detected in samples from poultry farms in Lijiang, Wenshan, and Yuxi. Evolutionary analysis and genotyping were performed for the 17 strains of isolated H9N2 virus. Phylogenetic analysis revealed that all H9N2 viral genes had 91.6%-100% nucleotide homology, belonged to the G57 genotype, and had high homology with H9N2 viruses isolated from Guangdong and Guangxi, suggesting that the H9N2 viruses in Yunnan Province may have been imported by chicks. Using a nucleotide divergence cutoff of 95%, we identified ten distinct H9N2 genotypes that continued to evolve. The surface genes of the H9N2 isolates displayed substantial genetic diversity, highlighting the genetic diversity and complexity of the H9N2-subtype AIVs in Yunnan. Molecular analysis demonstrated that all 17 strains of H9N2 isolates had mutations at H183N, Q226L, L31P, and I268V in hemagglutinin; S31N in matrix protein 2; and no replacements at positions 274 and 292 of the neuraminidase protein. Sixteen strains had the A558V mutation and one strain had the E627V mutation in polymerase basic protein 2. Analysis of these amino acid sites suggests that H9N2 influenza viruses in Yunnan continue to mutate and adapt to mammals and are sensitive to neuraminidase inhibitors but resistant to adamantanes. It is necessary to strengthen surveillance of AIV H9N2 subtypes in poultry and LPMs in Yunnan to further understand their genetic diversity.
Collapse
Affiliation(s)
- Xi Chen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Weiwu Mu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Yunteng Shao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Li Peng
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Rongjie Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Shiyu Luo
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Xingchen He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Limei Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Fengping He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Lei Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Ronghai Wang
- Animal Husbandry and Veterinary Bureau, Yanjin 657599, China
| | - Liangyu Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Bin Xiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
30
|
Wang S, Zhang Y, Zhou X, Ma Y, Shi J, Jiang Y, Li Y, Tian G, Wang X. Expression and characterization of recombinant antibodies against H7 subtype avian influenza virus and their diagnostic potential. Front Microbiol 2024; 15:1459402. [PMID: 39247689 PMCID: PMC11377330 DOI: 10.3389/fmicb.2024.1459402] [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: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Monoclonal antibodies (mAbs) play a pivotal role in disease diagnosis as well as immunotherapy interventions. Traditional monoclonal antibody generation relies on animal immunization procedures predominantly involving mice; however, recent advances in in-vitro expression methodologies have enabled large-scale production suitable for both industrial applications as well as scientific investigations. Methods In this study, two mAbs against H7 subtype avian influenza viruses (AIV) were sequenced and analyzed, and the DNA sequences encoding heavy chain (HC) and light chain (LC) were obtained and cloned into pCHO-1.0 expression vector. Then, the HC and LC expression plasmids were transfected into CHO-S cells to establish stable cell lines expressing these mAbs using a two-phase selection scheme with different concentrations of methotrexate and puromycin. Recombinant antibodies were purified from the cell culture medium, and their potential applications were evaluated using hemagglutination inhibition (HI), western blotting (WB), confocal microscopy, and enzyme-linked immunosorbent assay (ELISA). Results The results indicated that the obtained recombinant antibodies exhibited biological activity similar to that of the parent antibodies derived from ascites and could be used as a replacement for animal-derived mAbs. A kinetic analysis of the two antibodies to the AIV HA protein, conducted using surface plasmon resonance (SPR), showed concordance between the recombinant and parental antibodies. Discussion The data presented in this study suggest that the described antibody production protocol could avoid the use of experimental animals and better conform to animal welfare regulations, and provides a basis for further research and development of mAbs-based diagnostic products.
Collapse
Affiliation(s)
- Siwen Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ying Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- College of Life Engineering, Shenyang Institute of Technology, Shenyang, China
| | - Xu Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yue Ma
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yongping Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yanbing Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiurong Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
31
|
Jia X, Crawford JC, Gebregzabher D, Monson EA, Mettelman RC, Wan Y, Ren Y, Chou J, Novak T, McQuilten HA, Clarke M, Bachem A, Foo IJ, Fritzlar S, Carrera Montoya J, Trenerry AM, Nie S, Leeming MG, Nguyen THO, Kedzierski L, Littler DR, Kueh A, Cardamone T, Wong CY, Hensen L, Cabug A, Laguna JG, Agrawal M, Flerlage T, Boyd DF, Van de Velde LA, Habel JR, Loh L, Koay HF, van de Sandt CE, Konstantinov IE, Berzins SP, Flanagan KL, Wakim LM, Herold MJ, Green AM, Smallwood HS, Rossjohn J, Thwaites RS, Chiu C, Scott NE, Mackenzie JM, Bedoui S, Reading PC, Londrigan SL, Helbig KJ, Randolph AG, Thomas PG, Xu J, Wang Z, Chua BY, Kedzierska K. High expression of oleoyl-ACP hydrolase underpins life-threatening respiratory viral diseases. Cell 2024; 187:4586-4604.e20. [PMID: 39137778 DOI: 10.1016/j.cell.2024.07.026] [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: 07/07/2023] [Revised: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024]
Abstract
Respiratory infections cause significant morbidity and mortality, yet it is unclear why some individuals succumb to severe disease. In patients hospitalized with avian A(H7N9) influenza, we investigated early drivers underpinning fatal disease. Transcriptomics strongly linked oleoyl-acyl-carrier-protein (ACP) hydrolase (OLAH), an enzyme mediating fatty acid production, with fatal A(H7N9) early after hospital admission, persisting until death. Recovered patients had low OLAH expression throughout hospitalization. High OLAH levels were also detected in patients hospitalized with life-threatening seasonal influenza, COVID-19, respiratory syncytial virus (RSV), and multisystem inflammatory syndrome in children (MIS-C) but not during mild disease. In olah-/- mice, lethal influenza infection led to survival and mild disease as well as reduced lung viral loads, tissue damage, infection-driven pulmonary cell infiltration, and inflammation. This was underpinned by differential lipid droplet dynamics as well as reduced viral replication and virus-induced inflammation in macrophages. Supplementation of oleic acid, the main product of OLAH, increased influenza replication in macrophages and their inflammatory potential. Our findings define how the expression of OLAH drives life-threatening viral disease.
Collapse
Affiliation(s)
- Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Deborah Gebregzabher
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ebony A Monson
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yanmin Wan
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Yanqin Ren
- Shanghai Public Health Clinical Centre, Fudan University, Shanghai 201508, China
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Isabelle J Foo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Julio Carrera Montoya
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Alice M Trenerry
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Dene R Littler
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Andrew Kueh
- Walter Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Tina Cardamone
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Aira Cabug
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jaime Gómez Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group, University of Córdoba, International Excellence Agrifood Campus "CeiA3", 14014 Córdoba, Spain
| | - Mona Agrawal
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David F Boyd
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Lee-Ann Van de Velde
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Igor E Konstantinov
- Department of Cardiothoracic Surgery, Royal Children's Hospital, University of Melbourne, Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Parkville, VIC 3052, Australia
| | - Stuart P Berzins
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Katie L Flanagan
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS 7248, Australia; School of Health and Biomedical Science, RMIT University, Bundoora, VIC 3083, Australia; Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Marco J Herold
- Walter Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Amanda M Green
- Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Karla J Helbig
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Adrienne G Randolph
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA
| | - Jianqing Xu
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Zhongfang Wang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA.
| |
Collapse
|
32
|
Liu T, Liang L, Zhao P, Lin W, Zhuang Y, Jiang L, Chen H, Li C. The M2 Protein of the Influenza A Virus Interacts with PEX19 to Facilitate Virus Replication by Disrupting the Function of Peroxisome. Viruses 2024; 16:1309. [PMID: 39205283 PMCID: PMC11359511 DOI: 10.3390/v16081309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
The peroxisomal biogenesis factor 19 (PEX19) is necessary for early peroxisomal biogenesis. PEX19 has been implicated in the replication of a variety of viruses, but the details pertaining to the mechanisms of how PEX19 engages in the life cycle of these viruses still need to be elucidated. Here, we demonstrated that the C terminus of PEX19 interacted with the cytoplasmic tail region of the M2 protein of the influenza A virus (IAV) and inhibited the viral growth titers. IAV infection or PEX19 knockdown triggered a reduction in the peroxisome pool and led to the accumulation of ROS and cell damage, thereby creating favorable conditions for IAV replication. Moreover, a reduction in the peroxisome pool led to the attenuation of early antiviral response mediated by peroxisome MAVS and downstream type III interferons. This study also showed that the interaction between IAV M2 and PEX19 affected the binding of PEX19 to the peroxisome-associated protein PEX14 and peroxisome membrane protein 24 (PMP24). Collectively, our data demonstrate that host factor PEX19 suppresses the replication of the IAV, and the IAV employs its M2 protein to mitigate the restricting role of PEX19.
Collapse
Affiliation(s)
- Tanbin Liu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (T.L.); (P.Z.)
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| | - Libin Liang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China;
| | - Pu Zhao
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (T.L.); (P.Z.)
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| | - Weipeng Lin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| | - Yichao Zhuang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| | - Li Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| | - Hualan Chen
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (T.L.); (P.Z.)
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| | - Chengjun Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (W.L.); (Y.Z.); (L.J.)
| |
Collapse
|
33
|
Lefin N, Herrera-Belén L, Farias JG, Beltrán JF. Review and perspective on bioinformatics tools using machine learning and deep learning for predicting antiviral peptides. Mol Divers 2024; 28:2365-2374. [PMID: 37626205 DOI: 10.1007/s11030-023-10718-3] [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: 05/02/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023]
Abstract
Viruses constitute a constant threat to global health and have caused millions of human and animal deaths throughout human history. Despite advances in the discovery of antiviral compounds that help fight these pathogens, finding a solution to this problem continues to be a task that consumes time and financial resources. Currently, artificial intelligence (AI) has revolutionized many areas of the biological sciences, making it possible to decipher patterns in amino acid sequences that encode different functions and activities. Within the field of AI, machine learning, and deep learning algorithms have been used to discover antimicrobial peptides. Due to their effectiveness and specificity, antimicrobial peptides (AMPs) hold excellent promise for treating various infections caused by pathogens. Antiviral peptides (AVPs) are a specific type of AMPs that have activity against certain viruses. Unlike the research focused on the development of tools and methods for the prediction of antimicrobial peptides, those related to the prediction of AVPs are still scarce. Given the significance of AVPs as potential pharmaceutical options for human and animal health and the ongoing AI revolution, we have reviewed and summarized the current machine learning and deep learning-based tools and methods available for predicting these types of peptides.
Collapse
Affiliation(s)
- Nicolás Lefin
- Department of Chemical Engineering, Faculty of Engineering and Science, University of La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile
| | - Lisandra Herrera-Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomás, Temuco, Chile
| | - Jorge G Farias
- Department of Chemical Engineering, Faculty of Engineering and Science, University of La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile
| | - Jorge F Beltrán
- Department of Chemical Engineering, Faculty of Engineering and Science, University of La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile.
| |
Collapse
|
34
|
Zhai K, Dong J, Zeng J, Cheng P, Wu X, Han W, Chen Y, Qiu Z, Zhou Y, Pu J, Jiang T, Du X. Global antigenic landscape and vaccine recommendation strategy for low pathogenic avian influenza A (H9N2) viruses. J Infect 2024; 89:106199. [PMID: 38901571 DOI: 10.1016/j.jinf.2024.106199] [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: 12/19/2023] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
The sustained circulation of H9N2 avian influenza viruses (AIVs) poses a significant threat for contributing to a new pandemic. Given the temporal and spatial uncertainty in the antigenicity of H9N2 AIVs, the immune protection efficiency of vaccines remains challenging. By developing an antigenicity prediction method for H9N2 AIVs, named PREDAC-H9, the global antigenic landscape of H9N2 AIVs was mapped. PREDAC-H9 utilizes the XGBoost model with 14 well-designed features. The XGBoost model was built and evaluated to predict the antigenic relationship between any two viruses with high values of 81.1 %, 81.4 %, 81.3 %, 81.1 %, and 89.4 % in accuracy, precision, recall, F1 value, and area under curve (AUC), respectively. Then the antigenic correlation network (ACnet) was constructed based on the predicted antigenic relationship for H9N2 AIVs from 1966 to 2022, and ten major antigenic clusters were identified. Of these, four novel clusters were generated in China in the past decade, demonstrating the unique complex situation there. To help tackle this situation, we applied PREDAC-H9 to calculate the cluster-transition determining sites and screen out virus strains with the high cross-protective spectrum, thus providing an in silico reference for vaccine recommendation. The proposed model will reduce the clinical monitoring workload and provide a useful tool for surveillance and control of H9N2 AIVs.
Collapse
Affiliation(s)
- Ke Zhai
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jinze Dong
- National Key Laboratory of Veterinary Public Health and Safety, 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, PR China
| | - Jinfeng Zeng
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Peiwen Cheng
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xinsheng Wu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Wenjie Han
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yilin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Zekai Qiu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Department of Molecular and Radiooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Medical Faculty Heidelberg, Heidelberg University, Heidelberg 69047, Germany
| | - Yong Zhou
- National Key Laboratory of Veterinary Public Health and Safety, 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, PR China
| | - Juan Pu
- National Key Laboratory of Veterinary Public Health and Safety, 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, PR China.
| | - Taijiao Jiang
- Guangzhou National Laboratory, Guangzhou 510005, PR China; State Key Laboratory of Respiratory Disease, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China; Suzhou Institute of Systems Medicine, Suzhou 215123, PR China.
| | - Xiangjun Du
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Shenzhen Key Laboratory of Pathogenic Microbes & Biosecurity, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou 510030, PR China.
| |
Collapse
|
35
|
Ding X, Liu J, Jiang T, Wu A. Transmission restriction and genomic evolution co-shape the genetic diversity patterns of influenza A virus. Virol Sin 2024; 39:525-536. [PMID: 38423254 PMCID: PMC11401451 DOI: 10.1016/j.virs.2024.02.005] [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: 12/04/2023] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
Influenza A virus (IAV) shows an extensive host range and rapid genomic variations, leading to continuous emergence of novel viruses with significant antigenic variations and the potential for cross-species transmission. This causes global pandemics and seasonal flu outbreaks, posing sustained threats worldwide. Thus, studying all IAVs' evolutionary patterns and underlying mechanisms is crucial for effective prevention and control. We developed FluTyping to identify IAV genotypes, to explore overall genetic diversity patterns and their restriction factors. FluTyping groups isolates based on genetic distance and phylogenetic relationships using whole genomes, enabling identification of each isolate's genotype. Three distinct genetic diversity patterns were observed: one genotype domination pattern comprising only H1N1 and H3N2 seasonal influenza subtypes, multi-genotypes co-circulation pattern including majority avian influenza subtypes and swine influenza H1N2, and hybrid-circulation pattern involving H7N9 and three H5 subtypes of influenza viruses. Furthermore, the IAVs in multi-genotypes co-circulation pattern showed region-specific dominant genotypes, implying the restriction of virus transmission is a key factor contributing to distinct genetic diversity patterns, and the genomic evolution underlying different patterns was more influenced by host-specific factors. In summary, a comprehensive picture of the evolutionary patterns of overall IAVs is provided by the FluTyping's identified genotypes, offering important theoretical foundations for future prevention and control of these viruses.
Collapse
Affiliation(s)
- Xiao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, 100730, China
| | - Jingze Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, 100730, China
| | - Taijiao Jiang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, China; Guangzhou National Laboratory, Guangzhou, 510006, China; State Key Laboratory of Respiratory Disease, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510030, China.
| | - Aiping Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
36
|
Liu L, Wang F, Wu Y, Mi W, Zhang Y, Chen L, Wang D, Deng G, Shi J, Chen H, Kong H. The V223I substitution in hemagglutinin reduces the binding affinity to human-type receptors while enhancing the thermal stability of the H3N2 canine influenza virus. Front Microbiol 2024; 15:1442163. [PMID: 39104583 PMCID: PMC11299061 DOI: 10.3389/fmicb.2024.1442163] [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: 06/01/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Given the intimate relationship between humans and dogs, the H3N2 canine influenza viruses (CIVs) pose a threat to public health. In our study, we isolated four H3N2 CIVs from 3,758 dog nasal swabs in China between 2018 and 2020, followed by genetic and biological analysis. Phylogenetic analysis revealed 15 genotypes among all available H3N2 CIVs, with genotype 15 prevailing among dogs since around 2017, indicating the establishment of a stable virus lineage in dogs. Molecular characterization identified many mammalian adaptive substitutions, including HA-G146S, HA-N188D, PB2-I292T, PB2-G590S, PB2-S714I, PB1-D154G, and NP-R293K, present across the four isolates. Notably, analysis of HA sequences uncovered a newly emerged adaptive mutation, HA-V223I, which is predominantly found in human and swine H3N2 viruses, suggesting its role in mammalian adaptation. Receptor-binding analysis revealed that the four H3N2 viruses bind both avian and human-type receptors. However, HA-V223I decreases the H3N2 virus's affinity for human-type receptors but enhances its thermal stability. Furthermore, attachment analysis confirmed the H3N2 virus binding to human tracheal tissues, albeit with reduced affinity when the virus carries HA-V223I. Antigenic analysis indicated that the current human H3N2 vaccines do not confer protection against H3N2 CIVs. Collectively, these findings underscore that the potential threat posed by H3N2 CIVs to human health still exists, emphasizing the necessity of close surveillance and monitoring of H3N2 CIVs in dogs.
Collapse
Affiliation(s)
- Liling Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Fujun Wang
- Department of Biotechnology, Heilongjiang Vocational College for Nationalities, Harbin, China
- Harbin Fuai Pet Hospital, Harbin, China
| | - Ying Wu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Weiyong Mi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yaping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Lei Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Dongxue Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| |
Collapse
|
37
|
Yin Y, Zeng Z, Wei S, Shen Z, Cong Z, Zhu X. Using the sympathetic system, beta blockers and alpha-2 agonists, to address acute respiratory distress syndrome. Int Immunopharmacol 2024; 139:112670. [PMID: 39018694 DOI: 10.1016/j.intimp.2024.112670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) manifests as an acute inflammatory lung injury characterized by persistent hypoxemia, featuring a swift onset, high mortality, and predominantly supportive care as the current therapeutic approach, while effective treatments remain an area of active investigation. Adrenergic receptors (AR) play a pivotal role as stress hormone receptors, extensively participating in various inflammatory processes by initiating downstream signaling pathways. Advancements in molecular biology and pharmacology continually unveil the physiological significance of distinct AR subtypes. Interventions targeting these subtypes have the potential to induce specific alterations in cellular and organismal functions, presenting a promising avenue as a therapeutic target for managing ARDS. This article elucidates the pathogenesis of ARDS and the basic structure and function of AR. It also explores the relationship between AR and ARDS from the perspective of different AR subtypes, aiming to provide new insights for the improvement of ARDS.
Collapse
Affiliation(s)
- Yiyuan Yin
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Zhaojin Zeng
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Senhao Wei
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Ziyuan Shen
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhukai Cong
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China.
| | - Xi Zhu
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
38
|
Luo J, Zhang M, Ye Q, Gao F, Xu W, Li B, Wang Q, Zhao L, Tan WS. A synthetic TLR4 agonist significantly increases humoral immune responses and the protective ability of an MDCK-cell-derived inactivated H7N9 vaccine in mice. Arch Virol 2024; 169:163. [PMID: 38990396 DOI: 10.1007/s00705-024-06082-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024]
Abstract
Antigenically divergent H7N9 viruses pose a potential threat to public health, with the poor immunogenicity of candidate H7N9 vaccines demonstrated in clinical trials underscoring the urgent need for more-effective H7N9 vaccines. In the present study, mice were immunized with various doses of a suspended-MDCK-cell-derived inactivated H7N9 vaccine, which was based on a low-pathogenic H7N9 virus, to assess cross-reactive immunity and cross-protection against antigenically divergent H7N9 viruses. We found that the CRX-527 adjuvant, a synthetic TLR4 agonist, significantly enhanced the humoral immune responses of the suspended-MDCK-cell-derived H7N9 vaccine, with significant antigen-sparing and immune-enhancing effects, including robust virus-specific IgG, hemagglutination-inhibiting (HI), neuraminidase-inhibiting (NI), and virus-neutralizing (VN) antibody responses, which are crucial for protection against influenza virus infection. Moreover, the CRX-527-adjuvanted H7N9 vaccine also elicited cross-protective immunity and cross-protection against a highly pathogenic H7N9 virus with a single vaccination. Notably, NI and VN antibodies might play an important role in cross-protection against lethal influenza virus infections. This study showed that a synthetic TLR4 agonist adjuvant has a potent immunopotentiating effect, which might be considered worth further development as a means of increasing vaccine effectiveness.
Collapse
Affiliation(s)
- Jian Luo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Institute of Biological Products, Shanghai, China
| | - Min Zhang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Qian Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Feixia Gao
- Shanghai Institute of Biological Products, Shanghai, China
| | - Wenting Xu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Beibei Li
- Shanghai Institute of Biological Products, Shanghai, China
| | - Qi Wang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Liang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
39
|
Liang Z, Lin X, Sun L, Edwards KM, Song W, Sun H, Xie Y, Lin F, Ling S, Liang T, Xiao B, Wang J, Li M, Leung CY, Zhu H, Bhandari N, Varadarajan R, Levine MZ, Peiris M, Webster R, Dhanasekaran V, Leung NHL, Cowling BJ, Webby RJ, Ducatez M, Zanin M, Wong SS. A(H2N2) and A(H3N2) influenza pandemics elicited durable cross-reactive and protective antibodies against avian N2 neuraminidases. Nat Commun 2024; 15:5593. [PMID: 38961067 PMCID: PMC11222539 DOI: 10.1038/s41467-024-49884-9] [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: 08/22/2023] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
Human cases of avian influenza virus (AIV) infections are associated with an age-specific disease burden. As the influenza virus N2 neuraminidase (NA) gene was introduced from avian sources during the 1957 pandemic, we investigate the reactivity of N2 antibodies against A(H9N2) AIVs. Serosurvey of healthy individuals reveal the highest rates of AIV N2 antibodies in individuals aged ≥65 years. Exposure to the 1968 pandemic N2, but not recent N2, protected against A(H9N2) AIV challenge in female mice. In some older adults, infection with contemporary A(H3N2) virus could recall cross-reactive AIV NA antibodies, showing discernable human- or avian-NA type reactivity. Individuals born before 1957 have higher anti-AIV N2 titers compared to those born between 1957 and 1968. The anti-AIV N2 antibodies titers correlate with antibody titers to the 1957 N2, suggesting that exposure to the A(H2N2) virus contribute to this reactivity. These findings underscore the critical role of neuraminidase immunity in zoonotic and pandemic influenza risk assessment.
Collapse
Affiliation(s)
- Zaolan Liang
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xia Lin
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Lihong Sun
- Guangzhou Institute for Respiratory Health and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kimberly M Edwards
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wenjun Song
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, 510005, Guangdong Province, China
| | - Hailiang Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yanmin Xie
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Fangmei Lin
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Shiman Ling
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Tingting Liang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Biying Xiao
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jiaqi Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Min Li
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chin-Yu Leung
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, China
- Joint Institute of Virology (Shantou University and The University of Hong Kong), Guangdong-Hongkong Joint Laboratory of Emerging Infectious Diseases, Shantou University, Shantou, P. R. China
| | - Nisha Bhandari
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| | - Min Z Levine
- US Center for Disease Control and Prevention, Atlanta, GA, USA
| | - Malik Peiris
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Center for Immunology & Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Robert Webster
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Vijaykrishna Dhanasekaran
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Nancy H L Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Richard J Webby
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mariette Ducatez
- Interactions Hosts-Pathogens (IHAP), Université de Toulouse, National Research Institute for Agriculture, Food and the Environment (INRAE), National Veterinary School of Toulouse (ENVT), Toulouse, France
| | - Mark Zanin
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Center for Immunology & Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China.
| | - Sook-San Wong
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
40
|
Wang Y, Li Q, Peng P, Zhang Q, Huang Y, Hu J, Hu Z, Liu X. Dual N-linked glycosylation at residues 133 and 158 in the hemagglutinin are essential for the efficacy of H7N9 avian influenza virus like particle vaccine in chickens and mice. Vet Microbiol 2024; 294:110108. [PMID: 38729093 DOI: 10.1016/j.vetmic.2024.110108] [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: 11/27/2023] [Revised: 04/25/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
H7N9 subtype avian influenza virus (AIV) poses a great challenge to poultry industry. Virus-like particle (VLP) is a prospective alternative for the traditional egg-based influenza vaccines. N-linked glycosylation (NLG) regulates the efficacy of influenza vaccines, whereas the impact of NLG modifications on the efficacy of influenza VLP vaccines remains unclear. Here, H7N9 VLPs were assembled in insect cells through co-infection with the baculoviruses expressing the NLG-modified hemagglutinin (HA), neuraminidase and matrix proteins, and the VLP vaccines were assessed in chickens and mice. NLG modifications significantly enhanced hemagglutination-inhibition and virus neutralization antibody responses in mice, rather than in chickens, because different immunization strategies were used in these animal models. The presence of dual NLG at residues 133 and 158 significantly elevated HA-binding IgG titers in chickens and mice. The VLP vaccines conferred complete protection and significantly suppressed virus replication and lung pathology post challenge with H7N9 viruses in chickens and mice. VLP immunization activated T cell immunity-related cytokine response and inhibited inflammatory cytokine response in mouse lung. Of note, the presence of dual NLG at residues 133 and 158 optimized the capacity of the VLP vaccine to stimulate interleukin-4 expression, inhibit virus shedding or alleviate lung pathology in chickens or mice. Intriguingly, the VLP vaccine with NLG addition at residue 133 provided partial cross-protection against the H5Nx subtype AIVs in chickens and mice. In conclusion, dual NLG at residues 133 and 158 in HA can be potentially used to enhance the efficacy of H7N9 VLP vaccines in chickens and mammals.
Collapse
Affiliation(s)
- Yufei Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qun Li
- Yangzhou Uni-Bio Pharmaceutical Co., Ltd, Yangzhou, Jiangsu, China
| | - Peipei Peng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qi Zhang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yalan Huang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zenglei Hu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
41
|
Ni Z, Wang J, Yu X, Wang Y, Wang J, He X, Li C, Deng G, Shi J, Kong H, Jiang Y, Chen P, Zeng X, Tian G, Chen H, Bu Z. Influenza virus uses mGluR2 as an endocytic receptor to enter cells. Nat Microbiol 2024; 9:1764-1777. [PMID: 38849624 PMCID: PMC11222159 DOI: 10.1038/s41564-024-01713-x] [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: 11/14/2023] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Influenza virus infection is initiated by the attachment of the viral haemagglutinin (HA) protein to sialic acid receptors on the host cell surface. Most virus particles enter cells through clathrin-mediated endocytosis (CME). However, it is unclear how viral binding signals are transmitted through the plasma membrane triggering CME. Here we found that metabotropic glutamate receptor subtype 2 (mGluR2) and potassium calcium-activated channel subfamily M alpha 1 (KCa1.1) are involved in the initiation and completion of CME of influenza virus using an siRNA screen approach. Influenza virus HA directly interacted with mGluR2 and used it as an endocytic receptor to initiate CME. mGluR2 interacted and activated KCa1.1, leading to polymerization of F-actin, maturation of clathrin-coated pits and completion of the CME of influenza virus. Importantly, mGluR2-knockout mice were significantly more resistant to different influenza subtypes than the wild type. Therefore, blocking HA and mGluR2 interaction could be a promising host-directed antiviral strategy.
Collapse
Affiliation(s)
- Zixin Ni
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jinliang Wang
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xiaofei Yu
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yifan Wang
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jingfei Wang
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xijun He
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Chengjun Li
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Guohua Deng
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jianzhong Shi
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yongping Jiang
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Pucheng Chen
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xianying Zeng
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Guobin Tian
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Zhigao Bu
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| |
Collapse
|
42
|
James J, Thomas SS, Seekings AH, Mahmood S, Kelly M, Banyard AC, Núñez A, Brookes SM, Slomka MJ. Evaluating the epizootic and zoonotic threat of an H7N9 low-pathogenicity avian influenza virus (LPAIV) variant associated with enhanced pathogenicity in turkeys. J Gen Virol 2024; 105:002008. [PMID: 38980150 PMCID: PMC11316556 DOI: 10.1099/jgv.0.002008] [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: 01/22/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024] Open
Abstract
Between 2013 and 2017, the A/Anhui/1/13-lineage (H7N9) low-pathogenicity avian influenza virus (LPAIV) was epizootic in chickens in China, causing mild disease, with 616 fatal human cases. Despite poultry vaccination, H7N9 has not been eradicated. Previously, we demonstrated increased pathogenesis in turkeys infected with H7N9, correlating with the emergence of the L217Q (L226Q H3 numbering) polymorphism in the haemagglutinin (HA) protein. A Q217-containing virus also arose and is now dominant in China following vaccination. We compared infection and transmission of this Q217-containing 'turkey-adapted' (ty-ad) isolate alongside the H7N9 (L217) wild-type (wt) virus in different poultry species and investigated the zoonotic potential in the ferret model. Both wt and ty-ad viruses demonstrated similar shedding and transmission in turkeys and chickens. However, the ty-ad virus was significantly more pathogenic than the wt virus in turkeys but not in chickens, causing 100 and 33% mortality in turkeys respectively. Expanded tissue tropism was seen for the ty-ad virus in turkeys but not in chickens, yet the viral cell receptor distribution was broadly similar in the visceral organs of both species. The ty-ad virus required exogenous trypsin for in vitro replication yet had increased replication in primary avian cells. Replication was comparable in mammalian cells, and the ty-ad virus replicated successfully in ferrets. The L217Q polymorphism also affected antigenicity. Therefore, H7N9 infection in turkeys can generate novel variants with increased risk through altered pathogenicity and potential HA antigenic escape. These findings emphasize the requirement for enhanced surveillance and understanding of A/Anhui/1/13-lineage viruses and their risk to different species.
Collapse
Affiliation(s)
- Joe James
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
- WOAH/FAO International Reference Laboratory for Avian Influenza, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Saumya S. Thomas
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Amanda H. Seekings
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Sahar Mahmood
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Michael Kelly
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Ashley C. Banyard
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
- WOAH/FAO International Reference Laboratory for Avian Influenza, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Alejandro Núñez
- Pathology and Animal Sciences Department, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Sharon M. Brookes
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| | - Marek J. Slomka
- Department of Virology, Animal and Plant Health Agency (APHA-Weybridge), Woodham Lane, Addlestone, Surrey KT15 3NB, UK
| |
Collapse
|
43
|
Rostad CA, Atmar RL, Walter EB, Frey S, Meier JL, Sherman AC, Lai L, Tsong R, Kao CM, Raabe V, El Sahly HM, Keitel WA, Whitaker JA, Smith MJ, Schmader KE, Swamy GK, Abate G, Winokur P, Buchanan W, Cross K, Wegel A, Xu Y, Yildirim I, Kamidani S, Rouphael N, Roberts PC, Mulligan MJ, Anderson EJ. A Phase 2 Clinical Trial to Evaluate the Safety, Reactogenicity, and Immunogenicity of Different Prime-Boost Vaccination Schedules of 2013 and 2017 A(H7N9) Inactivated Influenza Virus Vaccines Administered With and Without AS03 Adjuvant in Healthy US Adults. Clin Infect Dis 2024; 78:1757-1768. [PMID: 38537255 PMCID: PMC11175706 DOI: 10.1093/cid/ciae173] [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: 01/23/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION A surge of human influenza A(H7N9) cases began in 2016 in China from an antigenically distinct lineage. Data are needed about the safety and immunogenicity of 2013 and 2017 A(H7N9) inactivated influenza vaccines (IIVs) and the effects of AS03 adjuvant, prime-boost interval, and priming effects of 2013 and 2017 A(H7N9) IIVs. METHODS Healthy adults (n = 180), ages 19-50 years, were enrolled into this partially blinded, randomized, multicenter phase 2 clinical trial. Participants were randomly assigned to 1 of 6 vaccination groups evaluating homologous versus heterologous prime-boost strategies with 2 different boost intervals (21 vs 120 days) and 2 dosages (3.75 or 15 μg of hemagglutinin) administered with or without AS03 adjuvant. Reactogenicity, safety, and immunogenicity measured by hemagglutination inhibition and neutralizing antibody titers were assessed. RESULTS Two doses of A(H7N9) IIV were well tolerated, and no safety issues were identified. Although most participants had injection site and systemic reactogenicity, these symptoms were mostly mild to moderate in severity; injection site reactogenicity was greater in vaccination groups receiving adjuvant. Immune responses were greater after an adjuvanted second dose, and with a longer interval between prime and boost. The highest hemagglutination inhibition geometric mean titer (95% confidence interval) observed against the 2017 A(H7N9) strain was 133.4 (83.6-212.6) among participants who received homologous, adjuvanted 3.75 µg + AS03/2017 doses with delayed boost interval. CONCLUSIONS Administering AS03 adjuvant with the second H7N9 IIV dose and extending the boost interval to 4 months resulted in higher peak antibody responses. These observations can broadly inform strategic approaches for pandemic preparedness. Clinical Trials Registration. NCT03589807.
Collapse
MESH Headings
- Humans
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/adverse effects
- Adult
- Male
- Female
- Middle Aged
- Influenza A Virus, H7N9 Subtype/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/adverse effects
- Antibodies, Viral/blood
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Young Adult
- Immunization, Secondary
- Immunization Schedule
- Hemagglutination Inhibition Tests
- United States
- Immunogenicity, Vaccine
- Antibodies, Neutralizing/blood
- Polysorbates/administration & dosage
- Polysorbates/adverse effects
- alpha-Tocopherol/administration & dosage
- alpha-Tocopherol/adverse effects
- Squalene/administration & dosage
- Squalene/adverse effects
- Squalene/immunology
- Healthy Volunteers
- Drug Combinations
- Adjuvants, Vaccine/administration & dosage
- Vaccination/methods
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/adverse effects
Collapse
Affiliation(s)
- Christina A Rostad
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Robert L Atmar
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Emmanuel B Walter
- Department of Pediatrics and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Sharon Frey
- Center for Vaccine Development, Saint Louis University, St. Louis, Missouri, USA
| | - Jeffery L Meier
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Amy C Sherman
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lilin Lai
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Carol M Kao
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Vanessa Raabe
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- New York University Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Hana M El Sahly
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wendy A Keitel
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jennifer A Whitaker
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael J Smith
- Department of Pediatrics and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Kenneth E Schmader
- Department of Medicine-Geriatrics, Duke University and GRECC, Durham VA Health Care System, Durham, North Carolina, USA
| | - Geeta K Swamy
- Department of Obstetrics and Gynecology and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Getahun Abate
- Center for Vaccine Development, Saint Louis University, St. Louis, Missouri, USA
| | - Patricia Winokur
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Wendy Buchanan
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | | | | | - Yongxian Xu
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Inci Yildirim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Satoshi Kamidani
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Nadine Rouphael
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Mark J Mulligan
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- New York University Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Evan J Anderson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
44
|
Jia M, Zhao H, Morano NC, Lu H, Lui YM, Du H, Becker JE, Yuen KY, Ho DD, Kwong PD, Shapiro L, To KKW, Wu X. Human neutralizing antibodies target a conserved lateral patch on H7N9 hemagglutinin head. Nat Commun 2024; 15:4505. [PMID: 38802413 PMCID: PMC11130183 DOI: 10.1038/s41467-024-48758-4] [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: 10/27/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Avian influenza A virus H7N9 causes severe human infections with >30% fatality. Currently, there is no H7N9-specific prevention or treatment for humans. Here, from a 2013 H7N9 convalescent case in Hong Kong, we isolate four hemagglutinin (HA)-reactive monoclonal antibodies (mAbs), with three directed to the globular head domain (HA1) and one to the stalk domain (HA2). Two clonally related HA1-directed mAbs, H7.HK1 and H7.HK2, potently neutralize H7N9 and protect female mice from lethal H7N9/AH1 challenge. Cryo-EM structures reveal that H7.HK1 and H7.HK2 bind to a β14-centered surface and disrupt the 220-loop that makes hydrophobic contacts with sialic acid on an adjacent protomer, thereby blocking viral entry. Sequence analysis indicates the lateral patch targeted by H7.HK1 and H7.HK2 to be conserved among influenza subtypes. Both H7.HK1 and H7.HK2 retain HA1 binding and neutralization capacity to later H7N9 isolates from 2016-2017, consistent with structural data showing that the antigenic mutations during this timeframe occur at their epitope peripheries. The HA2-directed mAb H7.HK4 lacks neutralizing activity but when used in combination with H7.HK2 moderately augments female mouse protection. Overall, our data reveal antibodies to a conserved lateral HA1 supersite that confer neutralization, and when combined with a HA2-directed non-neutralizing mAb, augment protection.
Collapse
Grants
- ZIA AI005022 Intramural NIH HHS
- W911NF-14-C-0001 U.S. Department of Defense (United States Department of Defense)
- FNIH SHAP19IUFV Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- Donations from Richard Yu and Carol Yu, Shaw Foundation Hong Kong, Michael Seak-Kan Tong, The Hui Ming, Hui Hoy and Chow Sin Lan Charity Fund Limited, Chan Yin Chuen Memorial Charitable Foundation, Marina Man-Wai Lee, Jessie and George Ho Charitable Foundation, Kai Chong Tong, Tse Kam Ming Laurence, Foo Oi Foundation Limited, Betty Hing-Chu Lee, and Ping Cham So
Collapse
Affiliation(s)
- Manxue Jia
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Hanjun Zhao
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, China
| | - Nicholas C Morano
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Department of Biochemistry, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
| | - Hong Lu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Yin-Ming Lui
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Haijuan Du
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jordan E Becker
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Department of Biochemistry, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, China
- Department of Clinical Microbiology and Infection, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Peter D Kwong
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lawrence Shapiro
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Department of Biochemistry, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, China.
- Department of Clinical Microbiology and Infection, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China.
| | - Xueling Wu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| |
Collapse
|
45
|
Su G, Chen Y, Li X, Shao JW. Virus versus host: influenza A virus circumvents the immune responses. Front Microbiol 2024; 15:1394510. [PMID: 38817972 PMCID: PMC11137263 DOI: 10.3389/fmicb.2024.1394510] [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: 03/01/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Influenza A virus (IAV) is a highly contagious pathogen causing dreadful losses to humans and animals around the globe. As is known, immune escape is a strategy that benefits the proliferation of IAVs by antagonizing, blocking, and suppressing immune surveillance. The HA protein binds to the sialic acid (SA) receptor to enter the cytoplasm and initiate viral infection. The conserved components of the viral genome produced during replication, known as the pathogen-associated molecular patterns (PAMPs), are thought to be critical factors for the activation of effective innate immunity by triggering dependent signaling pathways after recognition by pattern recognition receptors (PRRs), followed by a cascade of adaptive immunity. Viral infection-induced immune responses establish an antiviral state in the host to effectively inhibit virus replication and enhance viral clearance. However, IAV has evolved multiple mechanisms that allow it to synthesize and transport viral components by "playing games" with the host. At its heart, this review will describe how host and viral factors interact to facilitate the viral evasion of host immune responses.
Collapse
Affiliation(s)
- Guanming Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, China
| | - Yiqun Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Jian-Wei Shao
- School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
46
|
Cao N, Cai Y, Huang X, Jiang H, Huang Z, Xing L, Lu L, Jiang S, Xu W. Inhibition of influenza A virus and SARS-CoV-2 infection or co-infection by griffithsin and griffithsin-based bivalent entry inhibitor. mBio 2024; 15:e0074124. [PMID: 38587427 PMCID: PMC11077956 DOI: 10.1128/mbio.00741-24] [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: 03/10/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024] Open
Abstract
Outbreaks of acute respiratory viral diseases, such as influenza and COVID-19 caused by influenza A virus (IAV) and SARS-CoV-2, pose a serious threat to global public health, economic security, and social stability. This calls for the development of broad-spectrum antivirals to prevent or treat infection or co-infection of IAV and SARS-CoV-2. Hemagglutinin (HA) on IAV and spike (S) protein on SARS-CoV-2, which contain various types of glycans, play crucial roles in mediating viral entry into host cells. Therefore, they are key targets for the development of carbohydrate-binding protein-based antivirals. This study demonstrated that griffithsin (GRFT) and the GRFT-based bivalent entry inhibitor GL25E (GRFT-L25-EK1) showed broad-spectrum antiviral effects against IAV infection in vitro by binding to HA in a carbohydrate-dependent manner and effectively protected mice from lethal IAV infection. Although both GRFT and GL25E could inhibit infection of SARS-CoV-2 Omicron variants, GL25E proved to be significantly more effective than GRFT and EK1 alone. Furthermore, GL25E effectively inhibited in vitro co-infection of IAV and SARS-CoV-2 and demonstrated good druggability, including favorable safety and stability profiles. These findings suggest that GL25E is a promising candidate for further development as a broad-spectrum antiviral drug for the prevention and treatment of infection or co-infection from IAV and SARS-CoV-2.IMPORTANCEInfluenza and COVID-19 are highly contagious respiratory illnesses caused by the influenza A virus (IAV) and SARS-CoV-2, respectively. IAV and SARS-CoV-2 co-infection exacerbates damage to lung tissue and leads to more severe clinical symptoms, thus calling for the development of broad-spectrum antivirals for combating IAV and SARS-CoV-2 infection or co-infection. Here we found that griffithsin (GRFT), a carbohydrate-binding protein, and GL25E, a recombinant protein consisting of GRFT, a 25 amino acid linker, and EK1, a broad-spectrum coronavirus inhibitor, could effectively inhibit IAV and SARS-CoV-2 infection and co-infection by targeting glycans on HA of IAV and spike (S) protein of SARS-CoV-2. GL25E is more effective than GRFT because GL25E can also interact with the HR1 domain in SARS-CoV-2 S protein. Furthermore, GL25E possesses favorable safety and stability profiles, suggesting that it is a promising candidate for development as a drug to prevent and treat IAV and SARS-CoV-2 infection or co-infection.
Collapse
Affiliation(s)
- Najing Cao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanxing Cai
- Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou, China
| | - Xin Huang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hanxiao Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ziqi Huang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lixiao Xing
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Wang M, Liu Z, Liu C, He W, Qin D, You M. DNAzyme-based ultrasensitive immunoassay: Recent advances and emerging trends. Biosens Bioelectron 2024; 251:116122. [PMID: 38382271 DOI: 10.1016/j.bios.2024.116122] [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: 10/18/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 02/23/2024]
Abstract
Immunoassay, as the most commonly used method for protein detection, is simple to operate and highly specific. Sensitivity improvement is always the thrust of immunoassays, especially for the detection of trace quantities. The emergence of artificial enzyme, i.e., DNAzyme, provides a novel approach to improve the detection sensitivity of immunoassay. Simultaneously, its advantages of simple synthesis and high stability enable low cost, broad applicability and long shelf life for immunoassay. In this review, we summarized the recent advances in DNAzyme-based immunoassay. First, we summarized the existing different DNAzymes based on their catalytic activities. Next, the common signal amplification strategies used for DNAzyme-based immunoassays were reviewed to cater to diverse detection requirements. Following, the wide applications in disease diagnosis, environmental monitoring and food safety were discussed. Finally, the current challenges and perspectives on the future development of DNAzyme-based immunoassays were also provided.
Collapse
Affiliation(s)
- Meng Wang
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Zhe Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Chang Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Wanghong He
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, PR China
| | - Dui Qin
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China.
| | - Minli You
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China.
| |
Collapse
|
48
|
Yu DS, Wu XX, Weng TH, Cheng LF, Liu FM, Wu HB, Lu XY, Wu NP, Sun SL, Yao HP. Host proteins interact with viral elements and affect the life cycle of highly pathogenic avian influenza A virus H7N9. Heliyon 2024; 10:e28218. [PMID: 38560106 PMCID: PMC10981070 DOI: 10.1016/j.heliyon.2024.e28218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Host-virus interactions can significantly impact the viral life cycle and pathogenesis; however, our understanding of the specific host factors involved in highly pathogenic avian influenza A virus H7N9 (HPAI H7N9) infection is currently restricted. Herein, we designed and synthesized 65 small interfering RNAs targeting host genes potentially associated with various aspects of RNA virus life cycles. Afterward, HPAI H7N9 viruses were isolated and RNA interference was used to screen for host factors likely to be involved in the life cycle of HPAI H7N9. Moreover, the research entailed assessing the associations between host proteins and HPAI H7N9 proteins. Twelve key host proteins were identified: Annexin A (ANXA)2, ANXA5, adaptor related protein complex 2 subunit sigma 1 (AP2S1), adaptor related protein complex 3 subunit sigma 1 (AP3S1), ATP synthase F1 subunit alpha (ATP5A1), COPI coat complex subunit alpha (COP)A, COPG1, heat shock protein family A (Hsp70) member 1A (HSPA)1A, HSPA8, heat shock protein 90 alpha family class A member 1 (HSP90AA1), RAB11B, and RAB18. Co-immunoprecipitation revealed intricate interactions between viral proteins (hemagglutinin, matrix 1 protein, neuraminidase, nucleoprotein, polymerase basic 1, and polymerase basic 2) and these host proteins, presumably playing a crucial role in modulating the life cycle of HPAI H7N9. Notably, ANXA5, AP2S1, AP3S1, ATP5A1, HSP90A1, and RAB18, were identified as novel interactors with HPAI H7N9 proteins rather than other influenza A viruses (IAVs). These findings underscore the significance of host-viral protein interactions in shaping the dynamics of HPAI H7N9 infection, while highlighting subtle variations compared with other IAVs. Deeper understanding of these interactions holds promise to advance disease treatment and prevention strategies.
Collapse
Affiliation(s)
- Dong-Shan Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
- Department of Infectious Disease, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Xiao-Xin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Tian-Hao Weng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Lin-Fang Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Fu-Min Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Hai-Bo Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
- Department of Infectious Disease, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Xiang-Yun Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Nan-Ping Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, PR China
| | - Shui-Lin Sun
- Department of Infectious Disease, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, PR China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, PR China
| |
Collapse
|
49
|
He J, Deng J, Wen X, Yan M, Liu Y, Zhou Y, Du X, Yang H, Peng X. Isolation and genetic characteristics of Novel H4N1 Avian Influenza viruses in ChongQing, China. Virol J 2024; 21:85. [PMID: 38600529 PMCID: PMC11008002 DOI: 10.1186/s12985-024-02352-8] [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: 01/06/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Avian influenza viruses (AIVs) constitute significant zoonotic pathogens encompassing a broad spectrum of subtypes. Notably, the H4 subtype of AIVs has a pronounced ability to shift hosts. The escalating prevalence of the H4 subtype heightens the concern for its zoonotic potential, signaling an urgent need for vigilance. METHODS During the period from December 2021 to November 2023, we collected AIV-related environmental samples and assessed them using a comprehensive protocol that included nucleic acid testing, gene sequencing, isolation culture, and resequencing. RESULTS In this study, a total of 934 environmental samples were assessed, revealing a remarkably high detection rate (43.66%, 289/662) of AIV in the live poultry market. Notably, the H4N1 subtype AIV (cs2301) was isolated from the live poultry market and its complete genome sequence was successfully determined. Subsequent analysis revealed that cs2301, resulting from a reassortment event between wild and domesticated waterfowl, exhibits multiple mutations and demonstrates potential for host transfer. CONCLUSIONS Our research once again demonstrates the significant role of wild and domesticated waterfowl in the reassortment process of avian influenza virus, enriching the research on the H4 subtype of AIV, and emphasizing the importance of proactive monitoring the environment related to avian influenza virus.
Collapse
Affiliation(s)
- Jinyue He
- The affiliated Yongchuan hospital of Chongqing medical university, 402160, Yongchuan, China
| | - Jing Deng
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China.
| | - Xianxian Wen
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China
| | - Mengyuan Yan
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China
| | - Yang Liu
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China
| | - Yunqiu Zhou
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China
| | - XuBin Du
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China
| | - Han Yang
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China
| | - Xiaobin Peng
- Chongqing Changshou District Center for Disease Control and Prevention, 401220, Changshou, China.
| |
Collapse
|
50
|
Fang K, Song W, Zhang Y, Zheng Y, You C, Hu J, Liu L, Feng L, Zhao Z, Zhao Y, Wang J, Wang X, Zhu L, Chen T. Comparative analysis and prediction of avian influenza in Shangrao city, China from 2016 to 2022. Virology 2024; 592:109995. [PMID: 38290415 DOI: 10.1016/j.virol.2024.109995] [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: 12/24/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024]
Abstract
The aim of this study was to investigate the effects of vaccination, COVID-19 pandemic and migration of migratory birds on the avian influenza positivity rate in Shangrao City and to predict the future avian influenza positivity rate. Real-time reverse transcription polymerase chain reaction (RT-PCR) was used to detect nucleic acids of avian influenza A viruses. 1795 samples were collected between 2016 and 2022, of which 1086 were positive. In addition, there were seven human cases of avian influenza. The results showed that the positivity rate of H9 subtype in Shangrao City was higher than usual during the COVID-19 pandemic and migratory birds. Predictions suggest that the H9 subtype positivity rate in Shangrao City will be on the rise in the future. In recent years, the H5 positivity rate has gradually increased. Migratory birds and the COVID-19 pandemic have led to an increase in H9 subtype positivity. Therefore, the prevention and control of them should be strengthened.
Collapse
Affiliation(s)
- Kang Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, China
| | - Wentao Song
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, China
| | - Yanyan Zhang
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China
| | - Yiyang Zheng
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China
| | - Chen You
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China
| | - Jianhai Hu
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China
| | - Li Liu
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China
| | - Lei Feng
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China
| | - Zeyu Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, China
| | - Yunkang Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, China
| | - Jiayi Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, China
| | - Xiaolan Wang
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China; Shangrao People's Hospital, Shangrao City, Jiangxi Province, China.
| | - Lin Zhu
- Shangrao Center for Disease Control and Prevention, Shangrao City, Jiangxi Province, China.
| | - Tianmu Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, China.
| |
Collapse
|