1
|
Jin Z, Yi C, Zhou D, Wang X, Xie M, Zhou H, Zhang A. Chicken genome-wide CRISPR library screen identifies potential candidates associated with Avian influenza virus infection. Int J Biol Macromol 2025; 293:139267. [PMID: 39733882 DOI: 10.1016/j.ijbiomac.2024.139267] [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: 10/11/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
The avian influenza virus (AIV) poses a significant threat to both the poultry industry and public health. Systematic identification of host factors involved in AIV infection in chicken is critical. In this study, we developed a comprehensive chicken genome-wide sgRNA library containing 76,350 sgRNAs, with 4-6 sgRNAs designed per gene. Then, we constructed a genome-wide CRISPR/Cas9 knockout chicken fibroblasts cells (DF-1 cells) library, covering 99.9 % of the total sgRNAs. Following multiple rounds of survival selection during AIV infection, 706 potential genes were identified, including 107 genes previously associated with AIV infection. These candidate genes were primarily involved in ubiquitin-related pathways, RNA transport, endocytosis, and other cellular processes. Among these, 18 novel hits were selected and confirmed to contribute to AIV-induced cell death, with eight genes specifically implicated in AIV proliferation. Notably, RNF2 was found to negatively regulate interferon-stimulated genes (ISGs), DCP1A was suggested to influence gene expression linked to AIV proliferation, and CREB3L3 may regulate membrane cholesterol levels during AIV invasion, further validating the screening results. This study identified 599 potential chicken genes involved in AIV infection, providing a foundation for a deeper understanding of the mechanisms underlying AIV infection in avian cells.
Collapse
Affiliation(s)
- Zehua Jin
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Chenyang Yi
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Dongyu Zhou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Xiaoping Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Mengli Xie
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Hongbo Zhou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Anding Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, Hubei 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, Hubei 430070, China; Guangdong Provincial Key Laboratory of Research on the Technology of Pig-breeding and Pig-disease prevention, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
2
|
Rabezanahary H, Gilbert C, Santerre K, Scarrone M, Gilbert M, Thériault M, Brousseau N, Masson JF, Pelletier JN, Boudreau D, Trottier S, Baz M. Live virus neutralizing antibodies against pre and post Omicron strains in food and retail workers in Québec, Canada. Heliyon 2024; 10:e31026. [PMID: 38826717 PMCID: PMC11141348 DOI: 10.1016/j.heliyon.2024.e31026] [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: 10/30/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Background Measuring the ability of SARS-CoV-2 antibodies to neutralize live viruses remains an effective approach to quantify the level of protection of individuals. We assessed the neutralization activity against the ancestral SARS-CoV-2, Delta, Omicron BA.1, BA.2, BA.2.12.1, BA.4 and BA.5 strains, in 280 vaccinated restaurant/bar, grocery and hardware store workers in Québec, Canada. Methods Participants were recruited during the emergence of Omicron BA.1 variant. The neutralizing activity of participant sera was assessed by microneutralization assay. Results Serum neutralizing antibody (NtAb) titers of all participants against the ancestral SARS-CoV-2 strain were comparable with those against Delta variant (ranges of titers 10-2032 and 10-2560, respectively), however, their response was significantly reduced against Omicron BA.1, BA2, BA.2.12.1, BA.4 and BA.5 (10-1016, 10-1016, 10-320, 10-80 and 10-254, respectively). Individuals who received 2 doses of vaccine had significantly reduced NtAb titers against all SARS-CoV-2 strains compared to those infected and then vaccinated (≥1 dose), vaccinated (≥2 doses) and then infected, or those who received 3 doses of vaccine. Participants vaccinated with 2 or 3 doses of vaccine and then infected had the highest NtAb titers against all SARS-CoV-2 strains tested. Conclusion We assessed for the first time the NtAb response in food and retail workers. We found that vaccination prior to the emergence of Omicron BA.1 was associated with higher neutralizing activity against pre-Omicron variants, suggesting the importance of updating vaccines to increase antibody response against new SARS-CoV-2 variants. Vaccination followed by infection was associated with higher neutralizing activity against all SARS-CoV-2 strains tested.
Collapse
Affiliation(s)
- Henintsoa Rabezanahary
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Caroline Gilbert
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Kim Santerre
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Martina Scarrone
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Megan Gilbert
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Mathieu Thériault
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Nicholas Brousseau
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Biological Risks Department, Institut National de Santé Publique du Québec, Québec, QC, G1V 5B3, Canada
| | - Jean-François Masson
- Department of Chemistry, Quebec Center for Advanced Materials, Regroupement québécois sur les Matériaux de Pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Université de Montréal, Montréal, Canada
| | - Joelle N. Pelletier
- Department of Chemistry, Department of Biochemistry, Université de Montréal, Montréal, QC H2V 0B3, Canada
- PROTEO-The Québec Network for Research on Protein Function, Engineering, and Applications, Québec, Canada
| | - Denis Boudreau
- Département de Chimie et Centre d'Optique, Photonique et laser (COPL), Université Laval, Québec, Canada
| | - Sylvie Trottier
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Mariana Baz
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| |
Collapse
|
3
|
Kirk NM, Liang Y, Ly H. Comparative Pathology of Animal Models for Influenza A Virus Infection. Pathogens 2023; 13:35. [PMID: 38251342 PMCID: PMC10820042 DOI: 10.3390/pathogens13010035] [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: 10/18/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Animal models are essential for studying disease pathogenesis and to test the efficacy and safety of new vaccines and therapeutics. For most diseases, there is no single model that can recapitulate all features of the human condition, so it is vital to understand the advantages and disadvantages of each. The purpose of this review is to describe popular comparative animal models, including mice, ferrets, hamsters, and non-human primates (NHPs), that are being used to study clinical and pathological changes caused by influenza A virus infection with the aim to aid in appropriate model selection for disease modeling.
Collapse
Affiliation(s)
| | | | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN 55108, USA; (N.M.K.); (Y.L.)
| |
Collapse
|
4
|
Panzera Y, Mirazo S, Baz M, Techera C, Grecco S, Cancela F, Fuques E, Condon E, Calleros L, Camilo N, Fregossi A, Vaz I, Pessina P, Deshpande N, Pérez R, Benech A. Detection and genome characterisation of SARS-CoV-2 P.6 lineage in dogs and cats living with Uruguayan COVID-19 patients. Mem Inst Oswaldo Cruz 2023; 117:e220177. [PMID: 36651456 PMCID: PMC9870267 DOI: 10.1590/0074-02760220177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/07/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in domestic animals have occurred from the beginning of the pandemic to the present time. Therefore, from the perspective of One Health, investigating this topic is of global scientific and public interest. OBJECTIVES The present study aimed to determine the presence of SARS-CoV-2 in domestic animals whose owners had coronavirus disease 2019 (COVID-19). METHODS Nasopharyngeal and faecal samples were collected in Uruguay. Using quantitative polymerase chain reaction (qPCR), we analysed the presence of the SARS-CoV-2 genome. Complete genomes were obtained using ARTIC enrichment and Illumina sequencing. Sera samples were used for virus neutralisation assays. FINDINGS SARS-CoV-2 was detected in an asymptomatic dog and a cat. Viral genomes were identical and belonged to the P.6 Uruguayan SARS-CoV-2 lineage. Only antiserum from the infected cat contained neutralising antibodies against the ancestral SARS-CoV-2 strain and showed cross-reactivity against the Delta but not against the B.A.1 Omicron variant. MAIN CONCLUSIONS Domestic animals and the human SARS-CoV-2 P.6 variant comparison evidence a close relationship and gene flow between them. Different SARS-CoV-2 lineages infect dogs and cats, and no specific variants are adapted to domestic animals. This first record of SARS-CoV-2 in domestic animals from Uruguay supports regular surveillance of animals close to human hosts.
Collapse
Affiliation(s)
- Yanina Panzera
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay,+ Corresponding author:
| | - Santiago Mirazo
- Universidad de la República, Facultad de Ciencias, Sección Virología, Montevideo, Uruguay,Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Mariana Baz
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute, Melbourne, Victoria, Australia
| | - Claudia Techera
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay
| | - Sofía Grecco
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay
| | - Florencia Cancela
- Universidad de la República, Facultad de Ciencias, Sección Virología, Montevideo, Uruguay
| | - Eddie Fuques
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay
| | - Emma Condon
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay
| | - Lucía Calleros
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay
| | - Natalia Camilo
- Universidad de la República, Facultad de Veterinaria, Unidad de Clínica y Hospital Veterinario, Montevideo, Uruguay
| | - Andrea Fregossi
- Universidad de la República, Facultad de Veterinaria, Unidad de Clínica y Hospital Veterinario, Montevideo, Uruguay
| | - Inés Vaz
- Universidad de la República, Facultad de Veterinaria, Unidad de Clínica y Hospital Veterinario, Montevideo, Uruguay
| | - Paula Pessina
- Universidad de la República, Facultad de Veterinaria, Laboratorio Clínico del Hospital Veterinario, Montevideo, Uruguay
| | - Nikita Deshpande
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute, Melbourne, Victoria, Australia
| | - Ruben Pérez
- Universidad de la República, Facultad de Ciencias, Instituto de Biología, Departamento de Biología Animal, Sección Genética Evolutiva, Montevideo, Uruguay
| | - Alejandro Benech
- Universidad de la República, Facultad de Veterinaria, Unidad de Clínica y Hospital Veterinario, Montevideo, Uruguay
| |
Collapse
|
5
|
Vandoorn E, Stadejek W, Leroux-Roels I, Leroux-Roels G, Parys A, Van Reeth K. Human Immunity and Susceptibility to Influenza A(H3) Viruses of Avian, Equine, and Swine Origin. Emerg Infect Dis 2023; 29:98-109. [PMID: 36573615 PMCID: PMC9796212 DOI: 10.3201/eid2901.220943] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Influenza A viruses (IAVs) of subtype H3 that infect humans are antigenically divergent from those of birds, horses, and swine. Human immunity against these viruses might be limited, implying potential pandemic risk. To determine human risk, we selected 4 avian, 1 equine, and 3 swine IAVs representing major H3 lineages. We tested serum collected during 2017-2018 from 286 persons in Belgium for hemagglutination inhibiting antibodies and virus neutralizing antibodies against those animal-origin IAVs and tested replication in human airway epithelia. Seroprevalence rates for circulating IAVs from swine in North America were >51%, swine in Europe 7%-37%, and birds and equids ≤12%. Replication was efficient for cluster IV-A IAVs from swine in North America and IAVs from swine in Europe, intermediate for IAVs from horses and poultry, and absent for IAVs from wild birds and a novel human-like swine IAV in North America. Public health risk may be highest for swine H3 IAVs.
Collapse
|
6
|
Wang S, Xie Z, Huang L, Zhou X, Luo J, Yang Y, Li C, Duan P, Xu W, Chen D, Wu B, Yang Y, Liu X, Wang Y, Yuan Z, Qu D, Chen Z, Xia S. Safety and immunogenicity of an alum-adjuvanted whole-virion H7N9 influenza vaccine: a randomized, blinded, clinical trial. Clin Microbiol Infect 2020; 27:S1198-743X(20)30441-9. [PMID: 32738479 DOI: 10.1016/j.cmi.2020.07.033] [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: 02/03/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 11/18/2022]
Abstract
OBJECTIVES A case of H7N9 influenza virus infection was first identified in China in 2013. This virus is considered to have high pandemic potential. Here we developed an H7N9 influenza vaccine containing an aluminium adjuvant and evaluated the safety and immunogenicity of the vaccine. METHODS From October 2017 through August 2018 we conducted a randomized, double-blinded, single-centre phase I clinical trial in China among 360 participants aged ≥12 years. All participants received two doses of the vaccine (7.5, 15 or 30 μg haemagglutinin antigen) or placebo at an interval of 21 days. Adverse event data were collected for 30 days after vaccination. Serum samples were collected on days 0, 21 and 42 for the haemagglutinin inhibition (HI) antibody assay. RESULTS A total of 347 participants (347/360, 96.4%) completed the study. The proportions of vaccine-related adverse events after one injection were 56.7% (34/60) in the 7.5-μg group, 86.7% (52/60) in the 15-μg group and 86.7% (52/60) in the 30-μg group. The proportions of adverse events after two injections were less than those reported after the first dose. None of the serious adverse events were related to the vaccine. After receiving two doses of the 7.5-μg vaccine, the proportion of participants achieving an HI titre of ≥40 was 98.2% (55/56, 95%CI 72.3~100.0%), with a geometric mean titre (GMT) of 192.6 (95%CI 162.9~227.8). CONCLUSIONS The alum-adjuvanted H7N9 whole-virion inactivated vaccine was safe and strongly immunogenic in a population aged ≥12 years.
Collapse
Affiliation(s)
- Shilei Wang
- Shanghai Institute of Biological Products, Shanghai, China; Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhiqiang Xie
- Henan Province Centre for Disease Control and Prevention, Centre of Vaccine Clinical Research, China
| | - Lili Huang
- Henan Province Centre for Disease Control and Prevention, Centre of Vaccine Clinical Research, China
| | - Xu Zhou
- Shanghai Institute of Biological Products, Shanghai, China
| | - Jian Luo
- Shanghai Institute of Biological Products, Shanghai, China
| | - Yuelian Yang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Changgui Li
- National Institutes for Food and Drug Control, WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Beijing, China
| | - Peng Duan
- Shanghai Institute of Biological Products, Shanghai, China
| | - Wenting Xu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Dandan Chen
- Shanghai Institute of Biological Products, Shanghai, China
| | - Bing Wu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Yongli Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Xueying Liu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Yanxia Wang
- Henan Province Centre for Disease Control and Prevention, Centre of Vaccine Clinical Research, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Di Qu
- Biosafety Level-3 Laboratory, Key Laboratory of Medical Molecular Virology MOE & MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ze Chen
- Shanghai Institute of Biological Products, Shanghai, China.
| | - Shengli Xia
- Henan Province Centre for Disease Control and Prevention, Centre of Vaccine Clinical Research, China.
| |
Collapse
|
7
|
Belser JA, Pulit-Penaloza JA, Maines TR. Ferreting Out Influenza Virus Pathogenicity and Transmissibility: Past and Future Risk Assessments in the Ferret Model. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a038323. [PMID: 31871233 DOI: 10.1101/cshperspect.a038323] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
As influenza A viruses continue to jump species barriers, data generated in the ferret model to assess influenza virus pathogenicity, transmissibility, and tropism of these novel strains continues to inform an increasing scope of public health-based applications. This review presents the suitability of ferrets as a small mammalian model for influenza viruses and describes the breadth of pathogenicity and transmissibility profiles possible in this species following inoculation with a diverse range of viruses. Adaptation of aerobiology-based techniques and analyses have furthered our understanding of data obtained from this model and provide insight into the capacity of novel and emerging influenza viruses to cause human infection and disease.
Collapse
Affiliation(s)
- Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30329, USA
| | - Joanna A Pulit-Penaloza
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30329, USA
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30329, USA
| |
Collapse
|
8
|
Zou S, Tang J, Zhang Y, Liu L, Li X, Meng Y, Zhao X, Yang L, Shu Y, Wang D. Molecular characterization of H3 subtype avian influenza viruses based on poultry-related environmental surveillance in China between 2014 and 2017. Virology 2020; 542:8-19. [PMID: 31957664 DOI: 10.1016/j.virol.2020.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/26/2019] [Accepted: 01/04/2020] [Indexed: 12/17/2022]
Abstract
The H3 subtype avian influenza virus (AIV) poses a threat to both animal and human health. In this study, phylogenetic analysis showed that the H3 AIVs had various genomic constellations and extensive reassortments, increasing genetic diversity and the emergence of new pathogenic viruses that might infect human beings. Molecular analysis demonstrated that the major molecular markers linked to drug resistance were identified in M genes of three studied viruses, and there might be wide range of resistant virus infections in poultry in the future. Although all the H3 viruses preferentially bound to the avian-type receptor, the growth kinetics experiments showed that the selected H3 viruses were capable of efficient replication in mammalian cells, suggesting a potential cross-species transmission of H3 viruses. Overall, our results emphasize the need for continued surveillance of H3 outbreaks and may also help us improve knowledge on H3 AIVs prevention and control.
Collapse
Affiliation(s)
- Shumei Zou
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Jing Tang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Ye Zhang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Lijun Liu
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Xiyan Li
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Yao Meng
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Xiang Zhao
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Lei Yang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Yuelong Shu
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| | - Dayan Wang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| |
Collapse
|
9
|
Gahan J, Garvey M, Asmah Abd Samad R, Cullinane A. Whole Genome Sequencing of the First H3N8 Equine Influenza Virus Identified in Malaysia. Pathogens 2019; 8:E62. [PMID: 31083430 PMCID: PMC6630255 DOI: 10.3390/pathogens8020062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 01/23/2023] Open
Abstract
In August 2015, Malaysia experienced an outbreak of acute respiratory disease in racehorses. Clinical signs observed were consistent with equine influenza (EI) infection. The index cases were horses recently imported from New Zealand. Rapid control measures, including temporary cancellation of racing, were implemented to minimize the impact of the outbreak. By November, the disease outbreak was resolved, and movement restrictions were lifted. The aim of this study was to confirm the clinical diagnosis and characterize the causal virus. A pan-reactive influenza type A real-time RT-PCR was used for confirmatory diagnosis. Antigenic characterization by haemagglutinin inhibition using a panel of specific ferret antisera indicated that the causal virus belonged to clade 1 of the H3N8 Florida sub-lineage. The genetic characterization was achieved by the whole genome sequencing of positive nasal swabs from clinically affected animals. Pylogenetic analysis of the haemagglutinin (HA) and neuraminidase (NA) genes demonstrated ≥99% homology with several EI strains that had recently circulated in the USA and Japan. The antigenic and genetic characterization did not indicate that the current World Organisation for Animal Health (OIE) recommendations for EI vaccine composition required modification.
Collapse
Affiliation(s)
- Jacinta Gahan
- Virology Unit, The Irish Equine Centre, Johnstown, Naas, Co. Kildare W91 RH93, Ireland.
| | - Marie Garvey
- Virology Unit, The Irish Equine Centre, Johnstown, Naas, Co. Kildare W91 RH93, Ireland.
| | - Rozanah Asmah Abd Samad
- Department of Veterinary Services, Federal Government Administration Centre, 62630 Putrajaya, Malaysia.
| | - Ann Cullinane
- Virology Unit, The Irish Equine Centre, Johnstown, Naas, Co. Kildare W91 RH93, Ireland.
| |
Collapse
|
10
|
Bhide Y, Dong W, Gribonika I, Voshart D, Meijerhof T, de Vries-Idema J, Norley S, Guilfoyle K, Skeldon S, Engelhardt OG, Boon L, Christensen D, Lycke N, Huckriede A. Cross-Protective Potential and Protection-Relevant Immune Mechanisms of Whole Inactivated Influenza Virus Vaccines Are Determined by Adjuvants and Route of Immunization. Front Immunol 2019; 10:646. [PMID: 30984200 PMCID: PMC6450434 DOI: 10.3389/fimmu.2019.00646] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/11/2019] [Indexed: 12/27/2022] Open
Abstract
Adjuvanted whole inactivated virus (WIV) influenza vaccines show promise as broadly protective influenza vaccine candidates. Using WIV as basis we assessed the relative efficacy of different adjuvants by carrying out a head-to-head comparison of the liposome-based adjuvants CAF01 and CAF09 and the protein-based adjuvants CTA1-DD and CTA1-3M2e-DD and evaluated whether one or more of the adjuvants could induce broadly protective immunity. Mice were immunized with WIV prepared from A/Puerto Rico/8/34 (H1N1) virus intramuscularly with or without CAF01 or intranasally with or without CAF09, CTA1-DD, or CTA1-3M2e-DD, followed by challenge with homologous, heterologous or heterosubtypic virus. In general, intranasal immunizations were significantly more effective than intramuscular immunizations in inducing virus-specific serum-IgG, mucosal-IgA, and splenic IFNγ-producing CD4 T cells. Intranasal immunizations with adjuvanted vaccines afforded strong cross-protection with milder clinical symptoms and better control of virus load in lungs. Mechanistic studies indicated that non-neutralizing IgG antibodies and CD4 T cells were responsible for the improved cross-protection while IgA antibodies were dispensable. The role of CD4 T cells was particularly pronounced for CTA1-3M2e-DD adjuvanted vaccine as evidenced by CD4 T cell-dependent reduction of lung virus titers and clinical symptoms. Thus, intranasally administered WIV in combination with effective mucosal adjuvants appears to be a promising broadly protective influenza vaccine candidate.
Collapse
Affiliation(s)
- Yoshita Bhide
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wei Dong
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Inta Gribonika
- Department of Microbiology and Immunology, Institute of Biomedicine, Gothenburg University, Gothenburg, Sweden
| | - Daniëlle Voshart
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Tjarko Meijerhof
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jacqueline de Vries-Idema
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Stephen Norley
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Kate Guilfoyle
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), Medicines and Healthcare products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | - Sarah Skeldon
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), Medicines and Healthcare products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | - Othmar G Engelhardt
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), Medicines and Healthcare products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | | | - Dennis Christensen
- Adjuvant Research, Department of Infectious Diseases Immunology, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Nils Lycke
- Department of Microbiology and Immunology, Institute of Biomedicine, Gothenburg University, Gothenburg, Sweden
| | - Anke Huckriede
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
11
|
Fox A, Quinn KM, Subbarao K. Extending the Breadth of Influenza Vaccines: Status and Prospects for a Universal Vaccine. Drugs 2018; 78:1297-1308. [PMID: 30088204 DOI: 10.1007/s40265-018-0958-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the widespread use of seasonal influenza vaccines, there is urgent need for a universal influenza vaccine to provide broad, long-term protection. A number of factors underpin this urgency, including threats posed by zoonotic and pandemic influenza A viruses, suboptimal effectiveness of seasonal influenza vaccines, and concerns surrounding the effects of annual vaccination. In this article, we discuss approaches that are being investigated to increase influenza vaccine breadth, which are near-term, readily achievable approaches to increase the range of strains recognized within a subtype, or longer-term more challenging approaches to produce a truly universal influenza vaccine. Adjuvanted and neuraminidase-optimized vaccines are emerging as the most feasible and promising approaches to extend protection to cover a broader range of strains within a subtype. The goal of developing a universal vaccine has also been advanced with the design of immunogenic influenza HA-stem constructs that induce broadly neutralizing antibodies. However, these constructs are not yet sufficiently immunogenic to induce lasting universal immunity in humans. Advances in understanding how T cells mediate protection, and how viruses are packaged, have facilitated the rationale design and delivery of replication-incompetent virus vaccines that induce broad protection mediated by lung-resident memory T cells. While the lack of clear mechanistic correlates of protection, other than haemagglutination-inhibiting antibodies, remains an impediment to further advancing novel influenza vaccines, the pressing need for such a vaccine is supporting development of highly innovative and effective strategies.
Collapse
Affiliation(s)
- Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, and the Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, Australia
| | - Kylie M Quinn
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, and the Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, Australia.
| |
Collapse
|
12
|
Yang H, Xiao Y, Meng F, Sun F, Chen M, Cheng Z, Chen Y, Liu S, Chen H. Emergence of H3N8 equine influenza virus in donkeys in China in 2017. Vet Microbiol 2017; 214:1-6. [PMID: 29408020 DOI: 10.1016/j.vetmic.2017.11.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/26/2017] [Accepted: 11/26/2017] [Indexed: 11/26/2022]
Abstract
Equine influenza virus is a major respiratory pathogen in horses. Although both horses and donkeys belong to the genus Equus, donkey infection with influenza viruses is rare. In March 2017, an influenza outbreak occurred in donkeys in Shandong province, China. The causative virus, A/donkey/Shandong/1/2017(H3N8), was isolated from a dead donkey. Genetic analysis indicated that the virus originated from influenza A (H3N8) clade 2 of the Florida sub-lineage that has been circulating in Asian equine populations. Comparison of the deduced amino acid sequence of the HA gene of this causative virus with that of the A/equine/Richmond/1/2007 vaccine strain showed that substitutions had occurred in the antigenic regions A, B, and C. This study provides insight into the currently circulating and newly emerging H3N8 strains in donkeys in China.
Collapse
Affiliation(s)
- Huanliang Yang
- State Key Laboratory of Veterinary Biotechnology-Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yihong Xiao
- Department of Fundamental Veterinary Medicine, College of Animal Science and Veterinary Medicine-Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Shandong, China
| | - Fei Meng
- State Key Laboratory of Veterinary Biotechnology-Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Fachao Sun
- Department of Fundamental Veterinary Medicine, College of Animal Science and Veterinary Medicine-Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Shandong, China
| | - Meng Chen
- Department of Fundamental Veterinary Medicine, College of Animal Science and Veterinary Medicine-Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Shandong, China
| | - Zilong Cheng
- Department of Fundamental Veterinary Medicine, College of Animal Science and Veterinary Medicine-Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Shandong, China
| | - Yan Chen
- State Key Laboratory of Veterinary Biotechnology-Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Sidang Liu
- Department of Fundamental Veterinary Medicine, College of Animal Science and Veterinary Medicine-Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Shandong, China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology-Harbin Veterinary Research Institute, CAAS, Harbin, China.
| |
Collapse
|
13
|
Development of Clade-Specific and Broadly Reactive Live Attenuated Influenza Virus Vaccines against Rapidly Evolving H5 Subtype Viruses. J Virol 2017; 91:JVI.00547-17. [PMID: 28490598 DOI: 10.1128/jvi.00547-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/03/2017] [Indexed: 11/20/2022] Open
Abstract
We have developed pandemic live attenuated influenza vaccines (pLAIVs) against clade 1 H5N1 viruses on an Ann Arbor cold-adapted (ca) backbone that induced long-term immune memory. In 2015, many human infections caused by a new clade (clade 2.2.1.1) of goose/Guangdong (gs/GD) lineage H5N1 viruses were reported in Egypt, which prompted updating of the H5N1 pLAIV. We explored two strategies to generate suitable pLAIVs. The first approach was to modify the hemagglutinin gene of a highly pathogenic wild-type (wt) clade 2.2.1.1 virus, A/Egypt/N03434/2009 (Egy/09) (H5N1), with its unmodified neuraminidase (NA) gene; this virus was designated Egy/09 ca The second approach was to select a low-pathogenicity avian influenza H5 virus that elicited antibodies that cross-reacted with a broad range of H5 viruses, including the Egypt H5N1 viruses, and contained a novel NA subtype for humans. We selected the low-pathogenicity A/duck/Hokkaido/69/2000 (H5N3) (dk/Hok/00) virus for this purpose. Both candidate vaccines were attenuated and immunogenic in ferrets, inducing antibodies that neutralized homologous and heterologous H5 viruses with different degrees of cross-reactivity; Egy/09 ca vaccine antisera were more specific for the gs/GD lineage viruses but did not neutralize recent North American isolates (clade 2.3.4.4), whereas antisera from dk/Hok/69 ca-vaccinated ferrets cross-reacted with clade 2.3.4.4 and 2.2.1 viruses but not clade 1 or 2.1 viruses. When vaccinated ferrets were challenged with homologous and heterologous H5 viruses, challenge virus replication was reduced in the respiratory tract. Thus, the two H5 pLAIV candidates are suitable for clinical development to protect humans from infection with different clades of H5 viruses.IMPORTANCE In response to the continuing evolution of H5N1 avian influenza viruses and human infections, new candidate H5 live attenuated vaccines were developed by using two different approaches: one targeted a specific circulating strain in Egypt, and the other was based on a virus that elicits broadly cross-reactive antibodies against a wide range of H5 viruses. Both candidate vaccines were immunogenic and exhibited protective efficacy in ferrets. Our study permits a comparison of the two approaches, and the data support the further development of both vaccine viruses to optimally prepare for the further spread of clade 2.2.1 or 2.3.4.4 viruses.
Collapse
|
14
|
Safety and immunogenicity of an inactivated cell culture-derived H7N9 influenza vaccine in healthy adults: A phase I/II, prospective, randomized, open-label trial. Vaccine 2017; 35:4099-4104. [PMID: 28668573 DOI: 10.1016/j.vaccine.2017.06.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/26/2017] [Accepted: 06/19/2017] [Indexed: 01/27/2023]
Abstract
BACKGROUND We conducted a phase I/II clinical trial to evaluate the safety and immunogenicity of a Madin-Darby canine kidney (MDCK) cell-grown inactivated H7N9 influenza vaccine for pandemic preparedness purposes. METHODS Between April 7, 2015 and May 27, 2016, healthy adults aged 20-60years were enrolled sequentially in phase I (n=40) and phase II (n=160) from three hospitals in Taiwan and randomized to receive 2 doses of whole-virus H7N9 vaccine (15 or 30μg hemagglutinin antigen (HA) with or without an aluminum hydroxide adjuvant) at 21-day intervals. Safety up to 180days and changes in hemagglutinin inhibition (HI) titers at 21days after each vaccination were determined. RESULTS Of the 200 randomized subjects, 193 (96.5%) received 2 doses of the study vaccine and were included in the intention-to-treat analysis for safety, and 190 (95%) were included in the per-protocol analysis for immunogenicity. Most adverse events were mild and transient; no death or vaccine-related serious adverse events were reported. Overall, higher immune responses were observed in the groups administered with 30μgHA formulation than in the other two groups administered with 15μgHA formulation. The highest immune response was observed in subjects who received 2 doses of the adjuvanted vaccine containing 30μgHA with HI titer, seroprotection rate, seroconversion rate, and seroconversion factor of 36.2, 64.6%, 64.6% and 5.7, respectively. CONCLUSIONS Our study demonstrated that the H7N9 influenza vaccine containing 30µgHA with aluminum hydroxide adjuvant was immunogenic and safe in adults aged 20-60years. CLINICALTRIALS.GOV identifier: NCT02436928.
Collapse
|
15
|
Retamal M, Abed Y, Rhéaume C, Baz M, Boivin G. In vitro and in vivo evidence of a potential A(H1N1)pdm09 antigenic drift mediated by escape mutations in the haemagglutinin Sa antigenic site. J Gen Virol 2017. [PMID: 28631598 DOI: 10.1099/jgv.0.000800] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Influenza A(H1N1)pdm09 virus continues to circulate worldwide without evidence of significant antigenic drift between 2009 and 2016. By using escape mutants, we previously identified six haemagglutinin (HA) changes (T80R, G143E, G158E, N159D, K166E and A198E) that were located within antigenic sites. Combinations of these mutations were introduced into the A(H1N1)pdm09 HA plasmid by mutagenesis. Reassortant 6 : 2 viruses containing both the HA and NA genes of the A(H1N1)pdm09 and the six internal gene segments of A/PR/8/34 were rescued by reverse genetics. In vitro, HA inhibition and microneutralization assays showed that the HA hexa-mutant reassortant virus (RG1) escaped A(H1N1)pdm09 hyper-immune ferret antiserum recognition. C57Black/6 mice that received the vaccine formulated with A/California/07/09 were challenged with 2×104 p.f.u. of either the 6 : 2 wild-type (WT) or RG1 viruses. Reductions in body weight loss, mortality rate and lung viral titre were observed in immunized animals challenged with the 6 : 2 WT virus compared to non-immunized mice. However, immunization did not protect mice challenged with RG1 virus. To further characterize the mutations causing this antigenic change, 11 additional RG viruses whose HA gene contained single or combinations of mutations were evaluated in vitro. Although the RG1 virus was still the least reactive against hyper-immune serum by HAI testing, mutations G158E and N159D within the Sa antigenic site appeared to play the major role in the altered antigenicity of the A(H1N1)pdm09 virus. These results show that the Sa antigenic site contains the most prominent epitopes susceptible to cause an antigenic drift, escaping actual vaccine protection.
Collapse
Affiliation(s)
- Miguel Retamal
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, Québec City, Quebec, Canada
| | - Yacine Abed
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, Québec City, Quebec, Canada
| | - Chantal Rhéaume
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, Québec City, Quebec, Canada
| | - Mariana Baz
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, Québec City, Quebec, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, Québec City, Quebec, Canada
| |
Collapse
|
16
|
Pathogens Inactivated by Low-Energy-Electron Irradiation Maintain Antigenic Properties and Induce Protective Immune Responses. Viruses 2016; 8:v8110319. [PMID: 27886076 PMCID: PMC5127033 DOI: 10.3390/v8110319] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 12/26/2022] Open
Abstract
Inactivated vaccines are commonly produced by incubating pathogens with chemicals such as formaldehyde or β-propiolactone. This is a time-consuming process, the inactivation efficiency displays high variability and extensive downstream procedures are often required. Moreover, application of chemicals alters the antigenic components of the viruses or bacteria, resulting in reduced antibody specificity and therefore stimulation of a less effective immune response. An alternative method for inactivation of pathogens is ionizing radiation. It acts very fast and predominantly damages nucleic acids, conserving most of the antigenic structures. However, currently used irradiation technologies (mostly gamma-rays and high energy electrons) require large and complex shielding constructions to protect the environment from radioactivity or X-rays generated during the process. This excludes them from direct integration into biological production facilities. Here, low-energy electron irradiation (LEEI) is presented as an alternative inactivation method for pathogens in liquid solutions. LEEI can be used in normal laboratories, including good manufacturing practice (GMP)- or high biosafety level (BSL)-environments, as only minor shielding is necessary. We show that LEEI efficiently inactivates different viruses (influenza A (H3N8), porcine reproductive and respiratory syndrome virus (PRRSV), equine herpesvirus 1 (EHV-1)) and bacteria (Escherichia coli) and maintains their antigenicity. Moreover, LEEI-inactivated influenza A viruses elicit protective immune responses in animals, as analyzed by virus neutralization assays and viral load determination upon challenge. These results have implications for novel ways of developing and manufacturing inactivated vaccines with improved efficacy.
Collapse
|
17
|
Co-circulation of H5N6, H3N2, H3N8, and Emergence of Novel Reassortant H3N6 in a Local Community in Hunan Province in China. Sci Rep 2016; 6:25549. [PMID: 27151540 PMCID: PMC4858758 DOI: 10.1038/srep25549] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/19/2016] [Indexed: 12/15/2022] Open
Abstract
Multiple infections of avian influenza viruses (AIVs) in poultry or wild birds contribute to the continued evolution of H5 subtype viruses in nature and provide potential recombination of AIVs of different origins. In this study, we carried out surveillance of AIVs in ducks, geese and the environment of a community in Hunan province, China, from 2014–2015. We isolated multiple co-circulated AIVs including H3N2, H3N8, and H5N6, and, most importantly, a novel reassortant: H3N6. Phylogenetic analyses suggest that H3N6 is highly likely derived from H5N6, which has recently been shown to have zoonotic potential with human infections. Studies with mammalian cell lines and a mouse model indicate that four selected AIVs of duck or goose origin can infect MDCK and A549 cells but have low pathogenicity in mice. We propose that a potential co-circulation of multiple subtypes including H5N6 in local area may result in the production of novel subtypes such as H3N6 by gene reassortment.
Collapse
|
18
|
Gianchecchi E, Trombetta C, Piccirella S, Montomoli E. Evaluating influenza vaccines: progress and perspectives. Future Virol 2016. [DOI: 10.2217/fvl-2016-0012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Severe influenza infections are responsible for 3–5 million cases worldwide and 250,000–500,000 deaths per year. Although vaccination is the primary and most effective means of inducing protection against influenza viruses, it also presents limitations. This review outlines the promising steps that have been taken toward the development of a broadly protective influenza virus vaccine through the use of new technologies. The future challenge is to develop a broadly protective vaccine that is able to induce long-term protection against antigenically variant influenza viruses, regardless of antigenic shift and drift, and thus to protect against seasonal and pandemic influenza viruses.
Collapse
Affiliation(s)
- Elena Gianchecchi
- VisMederi Srl, Enterprise of Service in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy
| | - Claudia Trombetta
- Department of Molecular & Developmental Medicine, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Simona Piccirella
- VisMederi Srl, Enterprise of Service in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy
| | - Emanuele Montomoli
- VisMederi Srl, Enterprise of Service in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy
- Department of Molecular & Developmental Medicine, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| |
Collapse
|
19
|
Pavulraj S, Bera BC, Joshi A, Anand T, Virmani M, Vaid RK, Shanmugasundaram K, Gulati BR, Rajukumar K, Singh R, Misri J, Singh RK, Tripathi BN, Virmani N. Pathology of Equine Influenza virus (H3N8) in Murine Model. PLoS One 2015; 10:e0143094. [PMID: 26587990 PMCID: PMC4654517 DOI: 10.1371/journal.pone.0143094] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/30/2015] [Indexed: 01/09/2023] Open
Abstract
Equine influenza viruses (EIV)—H3N8 continue to circulate in equine population throughout the world. They evolve by the process of antigenic drift that leads to substantial change in the antigenicity of the virus, thereby necessitating substitution of virus strain in the vaccines. This requires frequent testing of the new vaccines in the in vivo system; however, lack of an appropriate laboratory animal challenge model for testing protective efficacy of equine influenza vaccine candidates hinders the screening of new vaccines and other therapeutic approaches. In the present investigation, BALB/c mouse were explored for suitability for conducting pathogenecity studies for EIV. The BALB/c mice were inoculated intranasally @ 2×106.24 EID50 with EIV (H3N8) belonging to Clade 2 of Florida sublineage and monitored for setting up of infection and associated parameters. All mice inoculated with EIV exhibited clinical signs viz. loss in body weights, lethargy, dyspnea, etc, between 3 and 5 days which commensurate with lesions observed in the respiratory tract including rhinitis, tracheitis, bronchitis, bronchiolitis, alveolitis and diffuse interstitial pneumonia. Transmission electron microscopy, immunohistochemistry, virus quantification through titration and qRT-PCR demonstrated active viral infection in the upper and lower respiratory tract. Serology revealed rise in serum lactate dehydrogenase levels along with sero-conversion. The pattern of disease progression, pathological lesions and virus recovery from nasal washings and lungs in the present investigations in mice were comparable to natural and experimental EIV infection in equines. The findings establish BALB/c mice as small animal model for studying EIV (H3N8) infection and will have immense potential for dissecting viral pathogenesis, vaccine efficacy studies, preliminary screening of vaccine candidates and antiviral therapeutics against EIV.
Collapse
Affiliation(s)
| | | | - Alok Joshi
- Veterinary Hospital—Naini, Barakot, Almora, Uttarakhand, India
| | - Taruna Anand
- National Research Centre on Equines, Hisar, Haryana, India
| | - Meenakshi Virmani
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary & Animal Sciences, Hisar, Haryana, India
| | | | | | | | - K. Rajukumar
- National Institute of High Security Animal Diseases, Bhopal, MP, India
| | - Rajendra Singh
- Division of Pathology, Indian Veterinary Research Institute, Bareilly, UP, India
| | - Jyoti Misri
- Division of Animal Science, Krishi Bhavan, New Delhi, India
| | | | | | - Nitin Virmani
- National Research Centre on Equines, Hisar, Haryana, India
- * E-mail:
| |
Collapse
|
20
|
Miller MA, Ganesan APV, Luckashenak N, Mendonca M, Eisenlohr LC. Endogenous antigen processing drives the primary CD4+ T cell response to influenza. Nat Med 2015; 21:1216-22. [PMID: 26413780 PMCID: PMC4629989 DOI: 10.1038/nm.3958] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 08/28/2015] [Indexed: 12/12/2022]
Abstract
By convention, CD4+ T lymphocytes recognize foreign and self peptides derived from internalized antigens in combination with MHC class II molecules. Alternative pathways of epitope production have been identified but their contributions to host defense have not been established. We show here in a mouse infection model that the CD4+ T cell response to influenza, critical for durable protection from the virus, is driven principally by unconventional processing of antigen synthesized within the infected antigen-presenting cell, not by classical processing of endocytosed virions or material from infected cells. Investigation of the cellular components involved, including the H2-M molecular chaperone, the proteasome, and gamma-interferon inducible lysosomal thiol reductase revealed considerable heterogeneity in the generation of individual epitopes, an arrangement that ensures peptide diversity and broad CD4+ T cell engagement. These results could fundamentally revise strategies for rational vaccine design and may lead to key insights into the induction of autoimmune and anti-tumor responses.
Collapse
Affiliation(s)
- Michael A Miller
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Asha Purnima V Ganesan
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nancy Luckashenak
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mark Mendonca
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Laurence C Eisenlohr
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Equine and Canine Influenza H3N8 Viruses Show Minimal Biological Differences Despite Phylogenetic Divergence. J Virol 2015; 89:6860-73. [PMID: 25903329 DOI: 10.1128/jvi.00521-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED The A/H3N8 canine influenza virus (CIV) emerged from A/H3N8 equine influenza virus (EIV) around the year 2000 through the transfer of a single virus from horses to dogs. We defined and compared the biological properties of EIV and CIV by examining their genetic variation, infection, and growth in different cell cultures, receptor specificity, hemagglutinin (HA) cleavage, and infection and growth in horse and dog tracheal explant cultures. Comparison of sequences of viruses from horses and dogs revealed mutations that may be linked to host adaptation and tropism. We prepared infectious clones of representative EIV and CIV strains that were similar to the consensus sequences of viruses from each host. The rescued viruses, including HA and neuraminidase (NA) double reassortants, exhibited similar degrees of long-term growth in MDCK cells. Different host cells showed various levels of susceptibility to infection, but no differences in infectivity were seen when comparing viruses. All viruses preferred α2-3- over α2-6-linked sialic acids for infections, and glycan microarray analysis showed that EIV and CIV HA-Fc fusion proteins bound only to α2-3-linked sialic acids. Cleavage assays showed that EIV and CIV HA proteins required trypsin for efficient cleavage, and no differences in cleavage efficiency were seen. Inoculation of the viruses into tracheal explants revealed similar levels of infection and replication by each virus in dog trachea, although EIV was more infectious in horse trachea than CIV. IMPORTANCE Influenza A viruses can cross species barriers and cause severe disease in their new hosts. Infections with highly pathogenic avian H5N1 virus and, more recently, avian H7N9 virus have resulted in high rates of lethality in humans. Unfortunately, our current understanding of how influenza viruses jump species barriers is limited. Our aim was to provide an overview and biological characterization of H3N8 equine and canine influenza viruses using various experimental approaches, since the canine virus emerged from horses approximately 15 years ago. We showed that although there were numerous genetic differences between the equine and canine viruses, this variation did not result in dramatic biological differences between the viruses from the two hosts, and the viruses appeared phenotypically equivalent in most assays we conducted. These findings suggest that the cross-species transmission and adaptation of influenza viruses may be mediated by subtle changes in virus biology.
Collapse
|
22
|
A Single Dose of an Avian H3N8 Influenza Virus Vaccine Is Highly Immunogenic and Efficacious against a Recently Emerged Seal Influenza Virus in Mice and Ferrets. J Virol 2015; 89:6907-17. [PMID: 25903333 DOI: 10.1128/jvi.00280-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 02/27/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED H3N8 influenza viruses are a commonly found subtype in wild birds, usually causing mild or no disease in infected birds. However, they have crossed the species barrier and have been associated with outbreaks in dogs, pigs, donkeys, and seals and therefore pose a threat to humans. A live attenuated, cold-adapted (ca) H3N8 vaccine virus was generated by reverse genetics using the wild-type (wt) hemagglutinin (HA) and neuraminidase (NA) genes from the A/blue-winged teal/Texas/Sg-00079/2007 (H3N8) (tl/TX/079/07) wt virus and the six internal protein gene segments from the ca influenza A virus vaccine donor strain, A/Ann Arbor/6/60 ca (H2N2), the backbone of the licensed seasonal live attenuated influenza vaccine. One dose of the tl/TX/079/07 ca vaccine induced a robust neutralizing antibody response against the homologous (tl/TX/079/07) and two heterologous influenza viruses, including the recently emerged A/harbor seal/New Hampshire/179629/2011 (H3N8) and A/northern pintail/Alaska/44228-129/2006 (H3N8) viruses, and conferred robust protection against the homologous and heterologous influenza viruses. We also analyzed human sera against the tl/TX/079/07 H3N8 avian influenza virus and observed low but detectable antibody reactivity in elderly subjects, suggesting that older H3N2 influenza viruses confer some cross-reactive antibody. The latter observation was confirmed in a ferret study. The safety, immunogenicity, and efficacy of the tl/TX/079/07 ca vaccine in mice and ferrets support further evaluation of this vaccine in humans for use in the event of transmission of an H3N8 avian influenza virus to humans. The human and ferret serology data suggest that a single dose of the vaccine may be sufficient in older subjects. IMPORTANCE Although natural infection of humans with an avian H3N8 influenza virus has not yet been reported, this influenza virus subtype has already crossed the species barrier and productively infected mammals. Pandemic preparedness is an important public health priority. Therefore, we generated a live attenuated avian H3N8 vaccine candidate and demonstrated that a single dose of the vaccine was highly immunogenic and protected mice and ferrets against homologous and heterologous H3N8 avian viruses.
Collapse
|
23
|
Molecular characterization of H9N2 influenza virus isolated from mink and its pathogenesis in mink. Vet Microbiol 2015; 176:88-96. [PMID: 25655813 DOI: 10.1016/j.vetmic.2015.01.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/24/2014] [Accepted: 01/08/2015] [Indexed: 01/15/2023]
Abstract
In mid-August 2013, two H9N2 influenza viruses, named A/mink/Shandong/F6/2013 (Mk/SD/F6/13) and A/mink/Shandong/F10/2013 (Mk/SD/F10/13), were isolated from lung samples of 2 of 45 farmed mink exhibiting respiratory signs in mideastern Shandong province, China. The seroprevalence of antibodies to H9N2 in mink was 20% (53/265). Based on sequence analysis, the eight nucleotide sequences showed 99.7-100% identity between Mk/SD/F6/13 and Mk/SD/F10/13. The HA, NP and NS genes of Mk/SD/F6/13 and Mk/SD/F10/13 were close to A/chicken/Zhejiang/329/2011 (H9N2), the NA and PB1 genes to A/duck/Hunan/S4111/2011 (H9N2), the PA and M genes to A/chicken/Shanghai/C1/2012 (H9N2). However, the PB2 genes had a close relationship with A/Turkey/California/189/66 (H9N2). Based on Sialic acid (SA) receptor detection, a range tissues of the mink demonstrated staining for MAA and/or SNA, and mink could serve as an intermediate host for influenza viruses with pandemic potential for the other animals. Experimental infection of mink demonstrated that mink could be infected by H9N2 influenza viruses and presented mild clinical signs, virus shedding and seroconversion, but no animals died of the disease. It implied that mammalian host-adapted avian H9N2 strains infected mink.
Collapse
|
24
|
A live attenuated equine H3N8 influenza vaccine is highly immunogenic and efficacious in mice and ferrets. J Virol 2014; 89:1652-9. [PMID: 25410860 DOI: 10.1128/jvi.02449-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Equine influenza viruses (EIV) are responsible for rapidly spreading outbreaks of respiratory disease in horses. Although natural infections of humans with EIV have not been reported, experimental inoculation of humans with these viruses can lead to a productive infection and elicit a neutralizing antibody response. Moreover, EIV have crossed the species barrier to infect dogs, pigs, and camels and therefore may also pose a threat to humans. Based on serologic cross-reactivity of H3N8 EIV from different lineages and sublineages, A/equine/Georgia/1/1981 (eq/GA/81) was selected to produce a live attenuated candidate vaccine by reverse genetics with the hemagglutinin and neuraminidase genes of the eq/GA/81 wild-type (wt) virus and the six internal protein genes of the cold-adapted (ca) A/Ann Arbor/6/60 (H2N2) vaccine donor virus, which is the backbone of the licensed seasonal live attenuated influenza vaccine. In both mice and ferrets, intranasal administration of a single dose of the eq/GA/81 ca vaccine virus induced neutralizing antibodies and conferred complete protection from homologous wt virus challenge in the upper respiratory tract. One dose of the eq/GA/81 ca vaccine also induced neutralizing antibodies and conferred complete protection in mice and nearly complete protection in ferrets upon heterologous challenge with the H3N8 (eq/Newmarket/03) wt virus. These data support further evaluation of the eq/GA/81 ca vaccine in humans for use in the event of transmission of an equine H3N8 influenza virus to humans. IMPORTANCE Equine influenza viruses have crossed the species barrier to infect other mammals such as dogs, pigs, and camels and therefore may also pose a threat to humans. We believe that it is important to develop vaccines against equine influenza viruses in the event that an EIV evolves, adapts, and spreads in humans, causing disease. We generated a live attenuated H3N8 vaccine candidate and demonstrated that the vaccine was immunogenic and protected mice and ferrets against homologous and heterologous EIV.
Collapse
|
25
|
Karlsson EA, Ip HS, Hall JS, Yoon SW, Johnson J, Beck MA, Webby RJ, Schultz-Cherry S. Respiratory transmission of an avian H3N8 influenza virus isolated from a harbour seal. Nat Commun 2014; 5:4791. [PMID: 25183346 PMCID: PMC4801029 DOI: 10.1038/ncomms5791] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 07/25/2014] [Indexed: 12/15/2022] Open
Abstract
The ongoing human H7N9 influenza infections highlight the threat of emerging avian influenza viruses. In 2011, an avian H3N8 influenza virus isolated from moribund New England harbour seals was shown to have naturally acquired mutations known to increase the transmissibility of highly pathogenic H5N1 influenza viruses. To elucidate the potential human health threat, here we evaluate a panel of avian H3N8 viruses and find that the harbour seal virus displays increased affinity for mammalian receptors, transmits via respiratory droplets in ferrets and replicates in human lung cells. Analysis of a panel of human sera for H3N8 neutralizing antibodies suggests that there is no population-wide immunity to these viruses. The prevalence of H3N8 viruses in birds and multiple mammalian species including recent isolations from pigs and evidence that it was a past human pandemic virus make the need for surveillance and risk analysis of these viruses of public health importance.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Base Sequence
- Birds
- Epithelial Cells/immunology
- Epithelial Cells/virology
- Ferrets
- Hemagglutinins, Viral/chemistry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/immunology
- Host Specificity
- Humans
- Immune Sera/chemistry
- Immunologic Surveillance
- Influenza A Virus, H3N8 Subtype/classification
- Influenza A Virus, H3N8 Subtype/genetics
- Influenza A Virus, H3N8 Subtype/immunology
- Influenza A Virus, H3N8 Subtype/pathogenicity
- Models, Molecular
- Molecular Sequence Data
- Orthomyxoviridae Infections/epidemiology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/transmission
- Orthomyxoviridae Infections/veterinary
- Phoca
- Phylogeny
- Protein Binding
- Receptors, Virus/chemistry
- Receptors, Virus/immunology
- Respiratory Mucosa/immunology
- Respiratory Mucosa/virology
- Respiratory System/immunology
- Respiratory System/virology
- Sialic Acids/chemistry
- Sialic Acids/immunology
- Swine
- United States/epidemiology
- Viral Tropism
Collapse
Affiliation(s)
- Erik A Karlsson
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Hon S Ip
- United States Geological Survey, National Wildlife Health Center, Madison, Wisconsin 53711, USA
| | - Jeffrey S Hall
- United States Geological Survey, National Wildlife Health Center, Madison, Wisconsin 53711, USA
| | - Sun Woo Yoon
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Jordan Johnson
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Melinda A Beck
- Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
26
|
Jang YH, Seong BL. Options and obstacles for designing a universal influenza vaccine. Viruses 2014; 6:3159-80. [PMID: 25196381 PMCID: PMC4147691 DOI: 10.3390/v6083159] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 12/13/2022] Open
Abstract
Since the discovery of antibodies specific to a highly conserved stalk region of the influenza virus hemagglutinin (HA), eliciting such antibodies has been considered the key to developing a universal influenza vaccine that confers broad-spectrum protection against various influenza subtypes. To achieve this goal, a prime/boost immunization strategy has been heralded to redirect host immune responses from the variable globular head domain to the conserved stalk domain of HA. While this approach has been successful in eliciting cross-reactive antibodies against the HA stalk domain, protective efficacy remains relatively poor due to the low immunogenicity of the domain, and the cross-reactivity was only within the same group, rather than among different groups. Additionally, concerns are raised on the possibility of vaccine-associated enhancement of viral infection and whether multiple boost immunization protocols would be considered practical from a clinical standpoint. Live attenuated vaccine hitherto remains unexplored, but is expected to serve as an alternative approach, considering its superior cross-reactivity. This review summarizes recent advancements in the HA stalk-based universal influenza vaccines, discusses the pros and cons of these approaches with respect to the potentially beneficial and harmful effects of neutralizing and non-neutralizing antibodies, and suggests future guidelines towards the design of a truly protective universal influenza vaccine.
Collapse
Affiliation(s)
- Yo Han Jang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, South Korea.
| | - Baik Lin Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, South Korea.
| |
Collapse
|
27
|
Hemagglutinin stalk-based universal vaccine constructs protect against group 2 influenza A viruses. J Virol 2013; 87:10435-46. [PMID: 23903831 DOI: 10.1128/jvi.01715-13] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Current influenza virus vaccines contain H1N1 (phylogenetic group 1 hemagglutinin), H3N2 (phylogenetic group 2 hemagglutinin), and influenza B virus components. These vaccines induce good protection against closely matched strains by predominantly eliciting antibodies against the membrane distal globular head domain of their respective viral hemagglutinins. This domain, however, undergoes rapid antigenic drift, allowing the virus to escape neutralizing antibody responses. The membrane proximal stalk domain of the hemagglutinin is much more conserved compared to the head domain. In recent years, a growing collection of antibodies that neutralize a broad range of influenza virus strains and subtypes by binding to this domain has been isolated. Here, we demonstrate that a vaccination strategy based on the stalk domain of the H3 hemagglutinin (group 2) induces in mice broadly neutralizing anti-stalk antibodies that are highly cross-reactive to heterologous H3, H10, H14, H15, and H7 (derived from the novel Chinese H7N9 virus) hemagglutinins. Furthermore, we demonstrate that these antibodies confer broad protection against influenza viruses expressing various group 2 hemagglutinins, including an H7 subtype. Through passive transfer experiments, we show that the protection is mediated mainly by neutralizing antibodies against the stalk domain. Our data suggest that, in mice, a vaccine strategy based on the hemagglutinin stalk domain can protect against viruses expressing divergent group 2 hemagglutinins.
Collapse
|