1
|
Fraiha ALS, da Silva Santos BSÁ, Aguilar NR, Gallinari GC, de Mendonça Angelo ALP, Costa JMC, Correia PA, Faustino LP, de Souza Silva TB, Guedes RMC, Guedes MIMC, de Magalhães Vieira Machado A, Costa EA, Lobato ZIP. Immunization and challenge trials in a murine model using different inactivated recombinant vaccines against H1N1 swine influenza virus circulating in Brazil. Vaccine 2025; 45:126638. [PMID: 39709805 DOI: 10.1016/j.vaccine.2024.126638] [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: 05/06/2024] [Revised: 12/07/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
In Brazil, at least four lineages of influenza A virus circulate pig population: 2009 H1N1 flu pandemic (pH1N1), human-seasonal origin H3N2, H1N1 and H1N2 (huH1 lineages) viruses. Studies related to the occurrence of swine influenza A virus (SIAV) in Brazilian herds have been detecting an increase of occurrence of huH1 lineages. This study aimed to construct recombinant vaccines against the huH1N1 virus and test the immunogens in a murine model. The virus was constructed by reverse genetics using plasmids encoding the HA and NA sequences from a wild huH1N1 virus isolated from an infected pig. Amplified virus was inactivated, and oil-in-water (OW) and gel polymer (GP) adjuvants were used to formulate the vaccines. C57Bl6 mice received two doses with 3 weeks interval by the intramuscular route. Animals were randomly divided into 8 groups (G1-G8): G1 received OW vaccine and G2 PBS plus OW adjuvant; G3 received GP vaccine and G4 PBS plus GP adjuvant; G5 received the live virus by the intranasal route while G6 only PBS; G7 and G8 did not receive any treatment. Serum samples were collected before vaccination and after the first and second dose. Except for G8, three weeks post boost animals were challenged with a wild huH1N1 virus and observed for weight changes. After infection, bronchoalveolar lavage fluid (BALF) and lungs were collected from animals of each group for viral titers and immunohistochemistry (IHC) analysis, respectively. After booster, vaccinated groups seroconverted and the vaccines induced protection upon challenge. Reverse Genetics technique can be used to produce new and quickly updated swine influenza vaccines which is promising to control the virus in Brazilian herds. Future studies may focus on using the technology to produce multivalent recombinant vaccines against distinct strains of SIAVs circulating in Brazilian pig herds.
Collapse
MESH Headings
- Animals
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Brazil
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/veterinary
- Orthomyxoviridae Infections/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Mice
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Swine
- Antibodies, Viral/blood
- Female
- Mice, Inbred C57BL
- Disease Models, Animal
- Adjuvants, Immunologic/administration & dosage
- Injections, Intramuscular
- Lung/virology
- Swine Diseases/prevention & control
- Adjuvants, Vaccine/administration & dosage
Collapse
Affiliation(s)
- Ana Luiza Soares Fraiha
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Nágila Rocha Aguilar
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Grazielle Cossenzo Gallinari
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Julia Machado Caetano Costa
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Paula Angélica Correia
- Department of Veterinary Clinic and Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lídia Paula Faustino
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, MG, Brazil
| | | | | | | | | | - Erica Azevedo Costa
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Zélia Inês Portela Lobato
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
2
|
De Meyer A, Meuleman P. Preclinical animal models to evaluate therapeutic antiviral antibodies. Antiviral Res 2024; 225:105843. [PMID: 38548022 DOI: 10.1016/j.antiviral.2024.105843] [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: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 04/05/2024]
Abstract
Despite the availability of effective preventative vaccines and potent small-molecule antiviral drugs, effective non-toxic prophylactic and therapeutic measures are still lacking for many viruses. The use of monoclonal and polyclonal antibodies in an antiviral context could fill this gap and provide effective virus-specific medical interventions. In order to develop these therapeutic antibodies, preclinical animal models are of utmost importance. Due to the variability in viral pathogenesis, immunity and overall characteristics, the most representative animal model for human viral infection differs between virus species. Therefore, throughout the years researchers sought to find the ideal preclinical animal model for each virus. The most used animal models in preclinical research include rodents (mice, ferrets, …) and non-human primates (macaques, chimpanzee, ….). Currently, antibodies are tested for antiviral efficacy against a variety of viruses including different hepatitis viruses, human immunodeficiency virus (HIV), influenza viruses, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and rabies virus. This review provides an overview of the current knowledge about the preclinical animal models that are used for the evaluation of therapeutic antibodies for the abovementioned viruses.
Collapse
Affiliation(s)
- Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
3
|
Green M, Al-Humadi N. Preclinical Toxicology of Vaccines. A COMPREHENSIVE GUIDE TO TOXICOLOGY IN NONCLINICAL DRUG DEVELOPMENT 2024:849-876. [DOI: 10.1016/b978-0-323-85704-8.00003-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Anderson TK, Medina RA, Nelson MI. The Evolution of SARS-CoV-2 and Influenza A Virus at the Human–Animal Interface. GENETICS AND EVOLUTION OF INFECTIOUS DISEASES 2024:549-572. [DOI: 10.1016/b978-0-443-28818-0.00016-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
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
|
6
|
Zabel B, Mäkelä SM, Nedveck D, Hibberd AA, Yeung N, Latvala S, Lehtoranta L, Junnila J, Walters KB, Morovic W, Lehtinen MJ. The Effect of Bifidobacterium animalis subsp. lactis Bl-04 on Influenza A Virus Infection in Mice. Microorganisms 2023; 11:2582. [PMID: 37894240 PMCID: PMC10609243 DOI: 10.3390/microorganisms11102582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Influenza A virus infection is a major global disease requiring annual vaccination. Clinical studies indicate that certain probiotics may support immune function against influenza and other respiratory viruses, but direct molecular evidence is scarce. Here, mice were treated with a placebo or Bifidobacterium animalis subsp. lactis Bl-04 (Bl-04) orally via food (cereal) and also by gavage and exposed to Influenza A virus H1N1 (H1N1). The symptoms of the infection were observed, and tissues and digesta were collected for viral load RT-qPCR, transcriptomics, and microbiomics. The treatment decreased the viral load by 48% at day 3 post-infection in lungs and symptoms of infection at day 4 compared to placebo. Tissue transcriptomics showed differences between the Bl-04 and placebo groups in the genes in the Influenza A pathway in the intestine, blood, and lungs prior to and post-infection, but the results were inconclusive. Moreover, 16S rRNA gene profiling and qPCR showed the presence of Bl-04 in the intestine, but without major shifts in the microbiome. In conclusion, Bl-04 treatment may influence the host response against H1N1 in a murine challenge model; however, further studies are required to elucidate the mechanism of action.
Collapse
Affiliation(s)
- Bryan Zabel
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Sanna M Mäkelä
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Derek Nedveck
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Ashley A Hibberd
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Nicolas Yeung
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Sinikka Latvala
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Liisa Lehtoranta
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | | | - Kevin B Walters
- Department of Infectious Disease Research, Southern Research Institute, 431 Aviation Way, Frederick, MD 21701, USA
| | - Wesley Morovic
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Markus J Lehtinen
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| |
Collapse
|
7
|
Song Y, Song J, Kim S, Jang H, Kim H, Jeong B, Park N, Kim S, Yong D, Lim EK, Lee KG, Kang T, Im SG. Charge-shifting polyplex as a viral RNA extraction carrier for streamlined detection of infectious viruses. MATERIALS HORIZONS 2023; 10:4571-4580. [PMID: 37581348 DOI: 10.1039/d3mh00861d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
The recent outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the need for rapid, user-friendly nucleic acid testing that involves simple but efficient RNA extraction. Here, we present a charge-shifting polyplex as an RNA extraction carrier for advanced diagnosis of infectious viral diseases. The polyplex comprises poly(2-(dimethylamino) ethyl acrylate) (pDMAEA) electrostatically conjugated with RNA. The pDMAEA film can rapidly dissolve in the viral RNA solution, promoting immediate binding with RNA to form the polyplex, which enables the efficient capture of a substantial quantity of RNA. Subsequently, the captured RNA can be readily released by the quick hydrolysis of pDMAEA at the onset of quantitative reverse transcription-polymerase chain reaction (qRT-PCR), streamlining the entire process from RNA extraction to analysis. The developed method requires only 5 min of centrifugation and enables the detection of RNA in a one-pot setup. Moreover, the proposed method is fully compatible with high-speed qRT-PCR kits and can identify clinical samples within 1 h including the entire extraction to detection procedure. Indeed, the method successfully detected influenza viruses, SARS-CoV-2, and their delta and omicron variants in 260 clinical samples with a sensitivity of 99.4% and specificity of 98.9%. This rapid, user-friendly polyplex-based approach represents a significant breakthrough in molecular diagnostics.
Collapse
Affiliation(s)
- Younseong Song
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Jayeon Song
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Seongeun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Hyowon Jang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Hogi Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Booseok Jeong
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Nahyun Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Sunjoo Kim
- Department of Laboratory Medicine, Gyeongsang National University College of Medicine, 79 Gangnam-ro, Jinju-si, Gyeongsangnam-do 52727, Republic of Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Eun-Kyung Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
- School of Pharmacy, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Kyoung G Lee
- Division of Nano-Bio Sensors/Chips Development, National NanoFab Center (NNFC), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
- School of Pharmacy, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|
8
|
Zhang L, Li Y(H, Kibler K, Kraberger S, Varsani A, Turk J, Elmadbouly N, Aliskevich E, Spaccarelli L, Estifanos B, Enow J, Zanetti IR, Saldevar N, Lim E, Schlievert J, Browder K, Wilson A, Juan FA, Pinteric A, Garg A, Monder H, Saju R, Gisriel S, Jacobs B, Karr TL, Florsheim EB, Kumar V, Wallen J, Rahman M, McFadden G, Hogue BG, Lucas AR. Viral anti-inflammatory serpin reduces immuno-coagulopathic pathology in SARS-CoV-2 mouse models of infection. EMBO Mol Med 2023; 15:e17376. [PMID: 37534622 PMCID: PMC10493584 DOI: 10.15252/emmm.202317376] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 08/04/2023] Open
Abstract
SARS-CoV-2 acute respiratory distress syndrome (ARDS) induces uncontrolled lung inflammation and coagulopathy with high mortality. Anti-viral drugs and monoclonal antibodies reduce early COVID-19 severity, but treatments for late-stage immuno-thrombotic syndromes and long COVID are limited. Serine protease inhibitors (SERPINS) regulate activated proteases. The myxoma virus-derived Serp-1 protein is a secreted immunomodulatory serpin that targets activated thrombotic, thrombolytic, and complement proteases as a self-defense strategy to combat clearance. Serp-1 is effective in multiple animal models of inflammatory lung disease and vasculitis. Here, we describe systemic treatment with purified PEGylated Serp-1 as a therapy for immuno-coagulopathic complications during ARDS. Treatment with PEGSerp-1 in two mouse-adapted SARS-CoV-2 models in C57Bl/6 and BALB/c mice reduced lung and heart inflammation, with improved outcomes. PEGSerp-1 significantly reduced M1 macrophages in the lung and heart by modifying urokinase-type plasminogen activator receptor (uPAR), thrombotic proteases, and complement membrane attack complex (MAC). Sequential changes in gene expression for uPAR and serpins (complement and plasminogen inhibitors) were observed. PEGSerp-1 is a highly effective immune-modulator with therapeutic potential for severe viral ARDS, immuno-coagulopathic responses, and Long COVID.
Collapse
Affiliation(s)
- Liqiang Zhang
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Yize (Henry) Li
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
- School of Life SciencesArizona State UniversityTempeAZUSA
| | - Karen Kibler
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Simona Kraberger
- Center of Fundamental and Applied MicrobiomicsBiodesign Institute, Arizona State UniversityTempeAZUSA
| | - Arvind Varsani
- School of Life SciencesArizona State UniversityTempeAZUSA
- Center of Fundamental and Applied MicrobiomicsBiodesign Institute, Arizona State UniversityTempeAZUSA
- Center for Evolution and Medicine, School of Life SciencesArizona State UniversityTempeAZUSA
| | - Julie Turk
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Nora Elmadbouly
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Emily Aliskevich
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Laurel Spaccarelli
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Bereket Estifanos
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Junior Enow
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Isabela Rivabem Zanetti
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Nicholas Saldevar
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Efrem Lim
- School of Life SciencesArizona State UniversityTempeAZUSA
- Center of Fundamental and Applied MicrobiomicsBiodesign Institute, Arizona State UniversityTempeAZUSA
| | - Jessika Schlievert
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Kyle Browder
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Anjali Wilson
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Fernando Arcos Juan
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Aubrey Pinteric
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Aman Garg
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Henna Monder
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Rohan Saju
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Savanah Gisriel
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
- Departments of Pathology & Lab MedicineYale‐New Haven HospitalNew HavenCTUSA
| | - Bertram Jacobs
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
- School of Life SciencesArizona State UniversityTempeAZUSA
| | - Timothy L Karr
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
- Neurodegenerative Disease Research Center & Proteomics Center, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Esther Borges Florsheim
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
- School of Life SciencesArizona State UniversityTempeAZUSA
| | - Vivek Kumar
- New Jersey Institute of TechnologyNewarkNJUSA
| | | | - Masmudur Rahman
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Grant McFadden
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
- School of Life SciencesArizona State UniversityTempeAZUSA
| | - Brenda G Hogue
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
- School of Life SciencesArizona State UniversityTempeAZUSA
- Center for Applied Structural Discovery, Biodesign InstituteArizona State UniversityTempeAZUSA
| | - Alexandra R Lucas
- Center for Personalized Diagnostics, Biodesign InstituteArizona State UniversityTempeAZUSA
- Center of Immunotherapy, Vaccines and Virotherapy, Biodesign InstituteArizona State UniversityTempeAZUSA
| |
Collapse
|
9
|
Kieran TJ, DaSilva J, Stark TJ, York IA, Pappas C, Barnes JR, Maines TR, Belser JA. Influenza A Virus Multicycle Replication Yields Comparable Viral Population Emergence in Human Respiratory and Ocular Cell Types. Microbiol Spectr 2023; 11:e0116623. [PMID: 37404140 PMCID: PMC10433845 DOI: 10.1128/spectrum.01166-23] [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: 03/16/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
While primarily considered a respiratory pathogen, influenza A virus (IAV) is nonetheless capable of spreading to, and replicating in, numerous extrapulmonary tissues in humans. However, within-host assessments of genetic diversity during multicycle replication have been largely limited to respiratory tract tissues and specimens. As selective pressures can vary greatly between anatomical sites, there is a need to examine how measures of viral diversity may vary between influenza viruses exhibiting different tropisms in humans, as well as following influenza virus infection of cells derived from different organ systems. Here, we employed human primary tissue constructs emulative of the human airway or corneal surface, and we infected both with a panel of human- and avian-origin IAV, inclusive of H1 and H3 subtype human viruses and highly pathogenic H5 and H7 subtype viruses, which are associated with both respiratory disease and conjunctivitis following human infection. While both cell types supported productive replication of all viruses, airway-derived tissue constructs elicited greater induction of genes associated with antiviral responses than did corneal-derived constructs. We used next-generation sequencing to examine viral mutations and population diversity, utilizing several metrics. With few exceptions, generally comparable measures of viral diversity and mutational frequency were detected following homologous virus infection of both respiratory-origin and ocular-origin tissue constructs. Expansion of within-host assessments of genetic diversity to include IAV with atypical clinical presentations in humans or in extrapulmonary cell types can provide greater insight into understanding those features most prone to modulation in the context of viral tropism. IMPORTANCE Influenza A virus (IAV) can infect tissues both within and beyond the respiratory tract, leading to extrapulmonary complications, such as conjunctivitis or gastrointestinal disease. Selective pressures governing virus replication and induction of host responses can vary based on the anatomical site of infection, yet studies examining within-host assessments of genetic diversity are typically only conducted in cells derived from the respiratory tract. We examined the contribution of influenza virus tropism on these properties two different ways: by using IAV associated with different tropisms in humans, and by infecting human cell types from two different organ systems susceptible to IAV infection. Despite the diversity of cell types and viruses employed, we observed generally similar measures of viral diversity postinfection across all conditions tested; these findings nonetheless contribute to a greater understanding of the role tissue type contributes to the dynamics of virus evolution within a human host.
Collapse
Affiliation(s)
- Troy J. Kieran
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Juliana DaSilva
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Thomas J. Stark
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ian A. York
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Claudia Pappas
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - John R. Barnes
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Taronna R. Maines
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica A. Belser
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Tosheva II, Saygan KS, Mijnhardt SM, Russell CJ, Fraaij PLA, Herfst S. Hemagglutinin stability as a key determinant of influenza A virus transmission via air. Curr Opin Virol 2023; 61:101335. [PMID: 37307646 DOI: 10.1016/j.coviro.2023.101335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 06/14/2023]
Abstract
To cause pandemics, zoonotic respiratory viruses need to adapt to replication in and spread between humans, either via (indirect or direct) contact or through the air via droplets and aerosols. To render influenza A viruses transmissible via air, three phenotypic viral properties must change, of which receptor-binding specificity and polymerase activity have been well studied. However, the third adaptive property, hemagglutinin (HA) acid stability, is less understood. Recent studies show that there may be a correlation between HA acid stability and virus survival in the air, suggesting that a premature conformational change of HA, triggered by low pH in the airways or droplets, may render viruses noninfectious before they can reach a new host. We here summarize available data from (animal) studies on the impact of HA acid stability on airborne transmission and hypothesize that the transmissibility of other respiratory viruses may also be impacted by an acidic environment in the airways.
Collapse
Affiliation(s)
- Ilona I Tosheva
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kain S Saygan
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands; Pandemic and Disaster Preparedness Center, Delft, Rotterdam, the Netherlands
| | - Suzanne Ma Mijnhardt
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands; Pandemic and Disaster Preparedness Center, Delft, Rotterdam, the Netherlands
| | - Charles J Russell
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pieter LA Fraaij
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands; Pandemic and Disaster Preparedness Center, Delft, Rotterdam, the Netherlands; Department of Paediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sander Herfst
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands; Pandemic and Disaster Preparedness Center, Delft, Rotterdam, the Netherlands.
| |
Collapse
|
11
|
Liu M, van Kuppeveld FJM, de Haan CAM, de Vries E. Gradual adaptation of animal influenza A viruses to human-type sialic acid receptors. Curr Opin Virol 2023; 60:101314. [DOI: 10.1016/j.coviro.2023.101314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
|
12
|
Pushan SS, Samantaray M, Rajagopalan M, Ramaswamy A. Evolution of Indian Influenza A (H1N1) Hemagglutinin Strains: A Comparative Analysis of the Pandemic Californian HA Strain. Front Mol Biosci 2023; 10:1111869. [PMID: 37006623 PMCID: PMC10061220 DOI: 10.3389/fmolb.2023.1111869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
The need for a vaccine/inhibitor design has become inevitable concerning the emerging epidemic and pandemic viral infections, and the recent outbreak of the influenza A (H1N1) virus is one such example. From 2009 to 2018, India faced severe fatalities due to the outbreak of the influenza A (H1N1) virus. In this study, the potential features of reported Indian H1N1 strains are analyzed in comparison with their evolutionarily closest pandemic strain, A/California/04/2009. The focus is laid on one of its surface proteins, hemagglutinin (HA), which imparts a significant role in attacking the host cell surface and its entry. The extensive analysis performed, in comparison with the A/California/04/2009 strain, revealed significant point mutations in all Indian strains reported from 2009 to 2018. Due to these mutations, all Indian strains disclosed altered features at the sequence and structural levels, which are further presumed to be associated with their functional diversity as well. The mutations observed with the 2018 HA sequence such as S91R, S181T, S200P, I312V, K319T, I419M, and E523D might improve the fitness of the virus in a new host and environment. The higher fitness and decreased sequence similarity of mutated strains may compromise therapeutic efficacy. In particular, the mutations observed commonly, such as serine-to-threonine, alanine-to-threonine, and lysine-to-glutamine at various regions, alter the physico-chemical features of receptor-binding domains, N-glycosylation, and epitope-binding sites when compared with the reference strain. Such mutations render diversity among all Indian strains, and the structural and functional characterization of these strains becomes inevitable. In this study, we observed that mutational drift results in the alteration of the receptor-binding domain, the generation of new variant N-glycosylation along with novel epitope-binding sites, and modifications at the structural level. Eventually, the pressing need to develop potentially distinct next-generation therapeutic inhibitors against the HA strains of the Indian influenza A (H1N1) virus is also highlighted here.
Collapse
Affiliation(s)
- Shilpa Sri Pushan
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| | - Mahesh Samantaray
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| | - Muthukumaran Rajagopalan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India
| | - Amutha Ramaswamy
- Department of Bioinformatics, Pondicherry University, Puducherry, India
| |
Collapse
|
13
|
Filarial infections compromise influenza vaccination efficacy: Lessons from the mouse. Immunol Lett 2023; 255:62-66. [PMID: 36889363 DOI: 10.1016/j.imlet.2023.03.001] [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: 01/03/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
Helminth parasites infect more than a quarter of the human population and inflict significant changes to the immunological status of their hosts. Several human studies report impaired responses to vaccinations in helminth-infected individuals. Analysing the impact of helminth infections on the efficacy of influenza vaccinations in the mouse system helps to elucidate the underlying immunological processes. Concurrent infection with the parasitic nematode Litomosoides sigmodontis reduced the quantity and quality of antibody responses to vaccination against seasonal influenza in BALB/c and C57BL/6 mice. This led to impaired vaccination-induced protection against challenge infections with the human pathogenic 2009 pandemic H1N1 influenza A virus in helminth-infected mice. Impaired responses were also observed if vaccinations were performed after immune-driven or drug-induced clearance of a previous helminth infection. Mechanistically, the suppression was associated with a systemic and sustained expansion of IL-10-producing CD4+CD49b+LAG-3+ type 1 regulatory T cells and partially abrogated by in vivo blockade of the IL-10 receptor. In summary, these findings raise the concern that individuals in helminth-endemic areas may not always benefit from vaccinations, even in the absence of an acute and diagnosable helminth infection.
Collapse
|
14
|
Utility of Human In Vitro Data in Risk Assessments of Influenza A Virus Using the Ferret Model. J Virol 2023; 97:e0153622. [PMID: 36602361 PMCID: PMC9888249 DOI: 10.1128/jvi.01536-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
As influenza A viruses (IAV) continue to cross species barriers and cause human infection, the establishment of risk assessment rubrics has improved pandemic preparedness efforts. In vivo pathogenicity and transmissibility evaluations in the ferret model represent a critical component of this work. As the relative contribution of in vitro experimentation to these rubrics has not been closely examined, we sought to evaluate to what extent viral titer measurements over the course of in vitro infections are predictive or correlates of nasal wash and tissue measurements for IAV infections in vivo. We compiled data from ferrets inoculated with an extensive panel of over 50 human and zoonotic IAV (inclusive of swine-origin and high- and low-pathogenicity avian influenza viruses associated with human infection) under a consistent protocol, with all viruses concurrently tested in a human bronchial epithelial cell line (Calu-3). Viral titers in ferret nasal wash specimens and nasal turbinate tissue correlated positively with peak titer in Calu-3 cells, whereas additional phenotypic and molecular determinants of influenza virus virulence and transmissibility in ferrets varied in their association with in vitro viral titer measurements. Mathematical modeling was used to estimate more generalizable key replication kinetic parameters from raw in vitro viral titers, revealing commonalities between viral infection progression in vivo and in vitro. Meta-analyses inclusive of IAV that display a diverse range of phenotypes in ferrets, interpreted with mathematical modeling of viral kinetic parameters, can provide critical information supporting a more rigorous and appropriate contextualization of in vitro experiments toward pandemic preparedness. IMPORTANCE Both in vitro and in vivo models are employed for assessing the pandemic potential of novel and emerging influenza A viruses in laboratory settings, but systematic examinations of how well viral titer measurements obtained in vitro align with results from in vivo experimentation are not frequently performed. We show that certain viral titer measurements following infection of a human bronchial epithelial cell line are positively correlated with viral titers in specimens collected from virus-inoculated ferrets and employ mathematical modeling to identify commonalities between viral infection progression between both models. These analyses provide a necessary first step in enhanced interpretation and incorporation of in vitro-derived data in risk assessment activities and highlight the utility of employing mathematical modeling approaches to more closely examine features of virus replication not identifiable by experimental studies alone.
Collapse
|
15
|
Pulit-Penaloza JA, Brock N, Belser JA, Sun X, Pappas C, Tumpey TM, Maines TR. Kinetics and magnitude of viral RNA shedding as indicators for Influenza A virus transmissibility in ferrets. Commun Biol 2023; 6:90. [PMID: 36690690 PMCID: PMC9871019 DOI: 10.1038/s42003-023-04459-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/10/2023] [Indexed: 01/24/2023] Open
Abstract
The ferret transmission model is routinely used to evaluate the pandemic potential of newly emerging influenza A viruses. However, concurrent measurement of viral load in the air is typically not a component of such studies. To address this knowledge gap, we measured the levels of virus in ferret nasal washes as well as viral RNA emitted into the air for 14 diverse influenza viruses, encompassing human-, swine-, and avian-origin strains. Here we show that transmissible viruses display robust replication and fast release into the air. In contrast, poorly- and non-transmissible viruses show significantly reduced or delayed replication along with lower detection of airborne viral RNA at early time points post inoculation. These findings indicate that efficient ferret-to-ferret transmission via the air is directly associated with fast emission of virus-laden particles; as such, quantification of viral RNA in the air represents a useful addition to established assessments of new influenza virus strains.
Collapse
Affiliation(s)
- Joanna A Pulit-Penaloza
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Nicole Brock
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Xiangjie Sun
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Claudia Pappas
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Terrence M Tumpey
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
16
|
Zhu M, He J, Zeng H, Wang P, Zhu Y, Sun F, Huang X, Xu Y, Huang C, Chen J, Guo X, Zhou H, Wei Z, Ouyang K, Huang W, Chen Y. Synergistic HA and NS mutations enhanced the virulence of a mouse-adapted H1N1 influenza A virus. Vet Microbiol 2023; 276:109615. [PMID: 36481481 DOI: 10.1016/j.vetmic.2022.109615] [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: 09/01/2022] [Revised: 11/15/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
H1N1 reassortants between the swine Eurasian avian-like (EA) and H1N1 2009 pandemic (H1N1 pdm/09) viruses have been circulating stably in pig populations for more than ten years, and they may have contributed to increased human infections. Whether these H1N1 viruses acquire adaptive mutations to increase their pathogenicity towards a new host is unknown. To address this problem, mouse-adapted (MA) variants of swine-origin EA H1N1 influenza virus isolated from dogs (A/canine/Guangxi/LZ57/2015[LZ57-MA]) were generated by serial lung-to-lung passages in BALB/c mice. These exhibited greater virulence and replication capability than the wild-type virus (LZ57-WT). Of the six adaptive mutations, two were mapped to the ribonucleoprotein (RNP) complex (PB2-E578D and PA-T97I), two to hemagglutinin (HA-N198D and HA-A227E) and two to the non-structural protein 1 (NS1) and nuclear export protein (NS1-A53D and NEP-R42K, respectively). Reverse genetic substitution of the viral genes and mutation experiments demonstrated that the mutations in PA-T97I could enhance the polymerase activity, but a significant downregulation of activity was seen with PB2-E578D, which was consistent with a decrease in virulence. However, HA and NS, which are genes that act synergistically, were found to be determinants of virulence in mice. The reassortant viruses bearing HA mutations (N198D and A227E) were acquired during adaptation enhanced early-stage viral replication in mammalian cells. The single-point mutations in the NS genes had limited effects on virulence. Furthermore, a combination of HA (N198D and A227E) with NS(A53D) in the rLZ57-WT backbone resulted in efficient replication and a significant increase in virulence. The results suggest that these substitutions could compensate for the polymerase function and contribute to enhanced virulence, which highlights a major role for mutations in the HA and NS genes.
Collapse
Affiliation(s)
- Min Zhu
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Jianqiao He
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Hao Zeng
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Pingping Wang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yaohui Zhu
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Fanyuan Sun
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Xin Huang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yi Xu
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Chongqiang Huang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Jiancai Chen
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Xinyi Guo
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Huabo Zhou
- Huabo Pet Hospital, Nanning 530004, PR China
| | - Zuzhang Wei
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease control, Nanning 530004, PR China; Guangxi College and Universities Key Laboratory of Prevention and Control for Animal Disease, Nanning 530004, PR China
| | - Kang Ouyang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease control, Nanning 530004, PR China; Guangxi College and Universities Key Laboratory of Prevention and Control for Animal Disease, Nanning 530004, PR China
| | - Weijian Huang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease control, Nanning 530004, PR China; Huabo Pet Hospital, Nanning 530004, PR China
| | - Ying Chen
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease control, Nanning 530004, PR China; Guangxi College and Universities Key Laboratory of Prevention and Control for Animal Disease, Nanning 530004, PR China.
| |
Collapse
|
17
|
Sequential Transmission of Influenza Viruses in Ferrets Does Not Enhance Infectivity and Does Not Predict Transmissibility in Humans. mBio 2022; 13:e0254022. [PMID: 36300929 PMCID: PMC9765597 DOI: 10.1128/mbio.02540-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Airborne transmission in ferrets is a key component of pandemic risk assessment. However, some emerging avian influenza viruses transmit between ferrets but do not spread in humans. Therefore, we evaluated sequential rounds of airborne transmission as an approach to enhance the predictive accuracy of the ferret model. We reasoned that infection of ferrets via the respiratory route and onward transmission would more closely model transmission in humans. We hypothesized that pandemic and seasonal viruses would transmit efficiently over two rounds of transmission, while emerging avian viruses would fail to transmit in a second round. The 2009 pandemic H1N1 (pdm09) and seasonal H3N2 viruses were compared to avian-origin H7N9 and H3N8 viruses. Depending on the virus strain, transmission efficiency varied from 50 to 100% during the first round of transmission; the efficiency for each virus did not change during the second round, and viral replication kinetics in both rounds of transmission were similar. Both the H1N1pdm09 and H7N9 viruses acquired specific mutations during sequential transmission, while the H3N2 and H3N8 viruses did not; however, a global analysis of host-adaptive mutations revealed that minimal changes were associated with transmission of H1N1 and H3N2 viruses, while a greater number of changes occurred in the avian H3N8 and H7N9 viruses. Thus, influenza viruses that transmit in ferrets maintain their transmission efficiency through serial rounds of transmission. This answers the question of whether ferrets can propagate viruses through more than one round of airborne transmission and emphasizes that transmission in ferrets is necessary but not sufficient to infer transmissibility in humans. IMPORTANCE Airborne transmission in ferrets is used to gauge the pandemic potential of emerging influenza viruses; however, some emerging influenza viruses that transmit between ferrets do not spread between humans. Therefore, we evaluated sequential rounds of airborne transmission in ferrets as a strategy to enhance the predictive accuracy of the ferret model. Human influenza viruses transmitted efficiently (>83%) over two rounds of airborne transmission, demonstrating that, like humans, ferrets infected by the respiratory route can propagate the infection onward through the air. However, emerging avian influenza viruses with associated host-adaptive mutations also transmitted through sequential transmission. Thus, airborne transmission in ferrets is necessary but not sufficient to infer transmissibility in humans, and sequential transmission did not enhance pandemic risk assessment.
Collapse
|
18
|
Pulit-Penaloza JA, Brock N, Jones J, Belser JA, Jang Y, Sun X, Thor S, Pappas C, Zanders N, Tumpey TM, Davis CT, Maines TR. Pathogenesis and transmission of human seasonal and swine-origin A(H1) influenza viruses in the ferret model. Emerg Microbes Infect 2022; 11:1452-1459. [PMID: 35537045 PMCID: PMC9176692 DOI: 10.1080/22221751.2022.2076615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Influenza A viruses (IAVs) in the swine reservoir constantly evolve, resulting in expanding genetic and antigenic diversity of strains that occasionally cause infections in humans and pose a threat of emerging as a strain capable of human-to-human transmission. For these reasons, there is an ongoing need for surveillance and characterization of newly emerging strains to aid pandemic preparedness efforts, particularly for the selection of candidate vaccine viruses and conducting risk assessments. Here, we performed a parallel comparison of the pathogenesis and transmission of genetically and antigenically diverse swine-origin A(H1N1) variant (v) and A(H1N2)v, and human seasonal A(H1N1)pdm09 IAVs using the ferret model. Both groups of viruses were capable of replication in the ferret upper respiratory tract; however, variant viruses were more frequently isolated from the lower respiratory tract as compared to the human-adapted viruses. Regardless of virus origin, observed clinical signs of infection differed greatly between strains, with some viruses causing nasal discharge, sneezing and, in some instances, diarrhea in ferrets. The most striking difference between the viruses was the ability to transmit through the air. Human-adapted viruses were capable of airborne transmission between all ferret pairs. In contrast, only one out of the four tested variant viruses was able to transmit via the air as efficiently as the human-adapted viruses. Overall, this work highlights the need for sustained monitoring of emerging swine IAVs to identify strains of concern such as those that are antigenically different from vaccine strains and that possess adaptations required for efficient respiratory droplet transmission in mammals.
Collapse
Affiliation(s)
- Joanna A Pulit-Penaloza
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Nicole Brock
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Joyce Jones
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Jessica A Belser
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Yunho Jang
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Xiangjie Sun
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Sharmi Thor
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Claudia Pappas
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Natosha Zanders
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Terrence M Tumpey
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - C Todd Davis
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| | - Taronna R Maines
- Centers for Disease Control and Prevention, Influenza Division, National Center for Immunization and Respiratory Diseases, Atlanta, GA, USA
| |
Collapse
|
19
|
Chavda V, Bezbaruah R, Kalita T, Sarma A, Devi JR, Bania R, Apostolopoulos V. Variant influenza: connecting the missing dots. Expert Rev Anti Infect Ther 2022; 20:1567-1585. [PMID: 36346383 DOI: 10.1080/14787210.2022.2144231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND In June 2009, the World Health Organization declared a new pandemic, the 2009 swine influenza pandemic (swine flu). The symptoms of the swine flu pandemic causing strain were comparable to most of the symptoms noted by seasonal influenza. AREA COVERED Zoonotic viruses that caused the swine flu pandemic and its preventive measures. EXPERT OPINION As per Centers for Disease Control and Prevention (CDC), the clinical manifestations in humans produced by the 2009 H1N1 'swine flu' virus were equivalent to the manifestations caused by related flu strains. The H1N1 vaccination was the most successful prophylactic measure since it prevented the virus from spreading and reduced the intensity and consequences of the pandemic. Despite the availability of therapeutics, the ongoing evolution and appearance of new strains have made it difficult to develop effective vaccines and therapies. Currently, the CDC recommends yearly flu immunization for those aged 6 months and above. The lessons learned from the A/2009/H1N1 pandemic in 2009 indicated that readiness of mankind toward new illnesses caused by mutant viral subtypes that leap from animals to people must be maintained.
Collapse
Affiliation(s)
- Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Tutumoni Kalita
- Department of Pharmaceutical Chemistry, Regional College of Pharmaceutical Sciences, RIPT Group of Institution, Sonapur, Guwahati, India
| | - Anupam Sarma
- Department of Pharmaceutics, Girijananda Chowdhury Institute of Pharmaceutical Science, Hatkhowapara, Azara, Guwahati, Assam, India
| | - Juti Rani Devi
- NETES Institute of Pharmaceutical Science, Mirza, Guwahati, India
| | - Ratnali Bania
- Pratiksha Institute of Pharmaceutical Sciences, India
| | | |
Collapse
|
20
|
Abstract
The continued spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in humans necessitates evaluation of variants for enhanced virulence and transmission. We used the ferret model to perform a comparative analysis of four SARS-CoV-2 strains, including an early pandemic isolate from the United States (WA1), and representatives of the Alpha, Beta, and Delta lineages. While Beta virus was not capable of pronounced replication in ferrets, WA1, Alpha, and Delta viruses productively replicated in the ferret upper respiratory tract, despite causing only mild disease with no overt histopathological changes. Strain-specific transmissibility was observed; WA1 and Delta viruses transmitted in a direct contact setting, whereas Delta virus was also capable of limited airborne transmission. Viral RNA was shed in exhaled air particles from all inoculated animals but was highest for Delta virus. Prior infection with SARS-CoV-2 offered varied protection against reinfection with either homologous or heterologous variants. Notable genomic variants in the spike protein were most frequently detected following WA1 and Delta virus infection.
Collapse
|
21
|
Hamele CE, Russell AB, Heaton NS. In Vivo Profiling of Individual Multiciliated Cells during Acute Influenza A Virus Infection. J Virol 2022; 96:e0050522. [PMID: 35867557 PMCID: PMC9327675 DOI: 10.1128/jvi.00505-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/03/2022] [Indexed: 01/09/2023] Open
Abstract
Influenza virus infections are thought to be initiated in a small number of cells; however, the heterogeneity across the cellular responses of the epithelial cells during establishment of disease is incompletely understood. Here, we used an H1N1 influenza virus encoding a fluorescent reporter gene, a cell lineage-labeling transgenic mouse line, and single-cell RNA sequencing to explore the range of responses in a susceptible epithelial cell population during an acute influenza A virus (IAV) infection. Focusing on multiciliated cells, we identified a subpopulation that basally expresses interferon-stimulated genes (ISGs), which we hypothesize may be important for the early response to infection. We subsequently found that a population of infected ciliated cells produce most of the ciliated cell-derived inflammatory cytokines, and nearly all bystander ciliated cells induce a broadly antiviral state. From these data together, we propose that variable preexisting gene expression patterns in the initial cells targeted by the virus may ultimately affect the establishment of viral disease. IMPORTANCE Influenza A virus poses a significant threat to public health, and each year, millions of people in the United States alone are exposed to the virus. We do not currently, however, fully understand why some individuals clear the infection asymptomatically and others become severely ill. Understanding how these divergent phenotypes arise could eventually be leveraged to design therapeutics that prevent severe disease. As a first step toward understanding these different infection states, we used a technology that allowed us to determine how thousands of individual murine lung epithelial cells behaved before and during IAV infection. We found that small subsets of epithelial cells exhibited an antiviral state prior to infection, and similarly, some cells made high levels of inflammatory cytokines during infection. We propose that different ratios of these individual cellular responses may contribute to the broader antiviral state of the lung and may ultimately affect disease severity.
Collapse
Affiliation(s)
- Cait E. Hamele
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alistair B. Russell
- Division of Biological Sciences, University of California, San Diego, San Diego, California, USA
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
22
|
Abstract
Past pandemic influenza viruses with sustained human-to-human transmissibility have emerged from animal influenza viruses. Employment of experimental models to assess the pandemic risk of emerging zoonotic influenza viruses provides critical information supporting public health efforts. Ferret transmission experiments have been utilized to predict the human-to-human transmission potential of novel influenza viruses. However, small sample sizes and a lack of standardized protocols can introduce interlaboratory variability, complicating interpretation of transmission experimental data. To assess the range of variation in ferret transmission experiments, a global exercise was conducted by 11 laboratories using two common stock H1N1 influenza viruses with different transmission characteristics in ferrets. Parameters known to affect transmission were standardized, including the inoculation route, dose, and volume, as well as a strict 1:1 donor/contact ratio for respiratory droplet transmission. Additional host and environmental parameters likely to affect influenza transmission kinetics were monitored and analyzed. The overall transmission outcomes for both viruses across 11 laboratories were concordant, suggesting the robustness of the ferret model for zoonotic influenza risk assessment. Among environmental parameters that varied across laboratories, donor-to-contact airflow directionality was associated with increased transmissibility. To attain high confidence in identifying viruses with moderate to high transmissibility or low transmissibility under a smaller number of participating laboratories, our analyses support the notion that as few as three but as many as five laboratories, respectively, would need to independently perform viral transmission experiments with concordant results. This exercise facilitates the development of a more homogenous protocol for ferret transmission experiments that are employed for the purposes of risk assessment.
Collapse
|
23
|
Zhu MM, Niu BW, Liu LL, Yang H, Qin BY, Peng XH, Chen LX, Liu Y, Wang C, Ren XN, Xu CH, Zhou XH, Li F. Development of a humanized HLA-A30 transgenic mouse model. Animal Model Exp Med 2022; 5:350-361. [PMID: 35791899 PMCID: PMC9434587 DOI: 10.1002/ame2.12225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/10/2022] Open
Abstract
Background There are remarkable genetic differences between animal major histocompatibility complex (MHC) systems and the human leukocyte antigen (HLA) system. HLA transgenic humanized mouse model systems offer a much better method to study the HLA‐A‐related principal mechanisms for vaccine development and HLA‐A‐restricted responses against infection in human. Methods A recombinant gene encoding the chimeric HLA‐A30 monochain was constructed. This HHD molecule contains the following: α1‐α2 domains of HLA‐A30, α3 and cytoplasmic domains of H‐2Db, linked at its N‐terminus to the C‐terminus of human β2m by a 15‐amino‐acid peptide linker. The recombinant gene encoding the chimeric HLA‐A30 monochain cassette was introduced into bacterial artificial chromosome (BAC) CH502‐67J3 containing the HLA‐A01 gene locus by Red‐mediated homologous recombination. Modified BAC CH502‐67J3 was microinjected into the pronuclei of wild‐type mouse oocytes. This humanized mouse model was further used to assess the immune responses against influenza A virus (H1N1) pdm09 clinically isolated from human patients. Immune cell population, cytokine production, and histopathology in the lung were analyzed. Results We describe a novel human β2m‐HLA‐A30 (α1α2)‐H‐2Db (α3 transmembrane cytoplasmic) (HHD) monochain transgenic mouse strain, which contains the intact HLA‐A01 gene locus including 49 kb 5′‐UTR and 74 kb 3′‐UTR of HLA‐A01*01. Five transgenic lines integrated into the large genomic region of HLA‐A gene locus were obtained, and the robust expression of exogenous transgene was detected in various tissues from A30‐18# and A30‐19# lines encompassing the intact flanking sequences. Flow cytometry revealed that the introduction of a large genomic region in HLA‐A gene locus can influence the immune cell constitution in humanized mice. Pdm09 infection caused a similar immune response among HLA‐A30 Tg humanized mice and wild‐type mice, and induced the rapid increase of cytokines, including IFN‐γ, TNF‐α, and IL‐6, in both HLA‐A30 humanized Tg mice and wild‐type mice. The expression of HLA‐A30 transgene was dramatically promoted in tissues from A30‐9# line at 3 days post‐infection (dpi). Conclusions We established a promising preclinical research animal model of HLA‐A30 Tg humanized mouse, which could accelerate the identification of novel HLA‐A30‐restricted epitopes and vaccine development, and support the study of HLA‐A‐restricted responses against infection in humans.
Collapse
Affiliation(s)
- Meng-Min Zhu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Bo-Wen Niu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Ling-Ling Liu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Hua Yang
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Bo-Yin Qin
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Xiu-Hua Peng
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Li-Xiang Chen
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Yang Liu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Chao Wang
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Xiao-Nan Ren
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Chun-Hua Xu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Xiao-Hui Zhou
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Feng Li
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| |
Collapse
|
24
|
Wei X, Du W, Duca M, Yu G, de Vries E, de Haan CAM, Pieters RJ. Preventing Influenza A Virus Infection by Mixed Inhibition of Neuraminidase and Hemagglutinin by Divalent Inhibitors. J Med Chem 2022; 65:7312-7323. [PMID: 35549211 PMCID: PMC9150099 DOI: 10.1021/acs.jmedchem.2c00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Divalent inhibitors
of the neuraminidase enzyme (NA) of the Influenza
A virus were synthesized with vastly different spacers. The spacers
varied from 14 to 56 atoms and were relatively rigid by way of the
building blocks and their connection by CuAAC. As the ligand for these
constructs, a Δ4-β-d-glucoronide was
used, which can be prepared form N-acetyl glucosamine.
This ligand showed good NA inhibitory potency but with room for improvement
by multivalency enhancement. The synthesized compounds showed modest
potency enhancement in NA activity assays but a sizeable potency increase
in a 4-day cytopathic effect assay. The demonstration that the compounds
can also inhibit hemagglutinin in addition to NA may be the cause
of the enhancement.
Collapse
Affiliation(s)
- Xuan Wei
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, Utrecht NL-3508 TB, The Netherlands
| | - Wenjuan Du
- Section Virology, Division Infectious Diseases and Immunology, Faculty Veterinary Medicine, Utrecht University, Utrecht NL-3508 TB, The Netherlands
| | - Margherita Duca
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, Utrecht NL-3508 TB, The Netherlands
| | - Guangyun Yu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, Utrecht NL-3508 TB, The Netherlands
| | - Erik de Vries
- Section Virology, Division Infectious Diseases and Immunology, Faculty Veterinary Medicine, Utrecht University, Utrecht NL-3508 TB, The Netherlands
| | - Cornelis A M de Haan
- Section Virology, Division Infectious Diseases and Immunology, Faculty Veterinary Medicine, Utrecht University, Utrecht NL-3508 TB, The Netherlands
| | - Roland J Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, Utrecht NL-3508 TB, The Netherlands
| |
Collapse
|
25
|
Pre-existing helminth infection impairs the efficacy of adjuvanted influenza vaccination in mice. PLoS One 2022; 17:e0266456. [PMID: 35358281 PMCID: PMC8970517 DOI: 10.1371/journal.pone.0266456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 03/21/2022] [Indexed: 01/15/2023] Open
Abstract
The world health organization estimates that more than a quarter of the human population is infected with parasitic worms that are called helminths. Many helminths suppress the immune system of their hosts to prolong their survival. This helminth-induced immunosuppression “spills over” to unrelated antigens and can suppress the immune response to vaccination against other pathogens. Indeed, several human studies have reported a negative correlation between helminth infections and responses to vaccinations. Using mice that are infected with the parasitic nematode Litomosoides sigmodontis as a model for chronic human filarial infections, we reported previously that concurrent helminth infection impaired the vaccination-induced protection against the human pathogenic 2009 pandemic H1N1 influenza A virus (2009 pH1N1). Vaccinated, helminth-infected mice produced less neutralizing, influenza-specific antibodies than vaccinated naïve control mice. Consequently helminth-infected and vaccinated mice were not protected against a challenge infection with influenza virus but displayed high virus burden in the lung and a transient weight loss. In the current study we tried to improve the vaccination efficacy using vaccines that are licensed for humans. We either introduced a prime-boost vaccination regimen using the non-adjuvanted anti-influenza vaccine Begripal or employed the adjuvanted influenza vaccine Fluad. Although both strategies elevated the production of influenza-specific antibodies and protected mice from the transient weight loss that is caused by an influenza challenge infection, sterile immunity was not achieved. Helminth-infected vaccinated mice still had high virus burden in the lung while non-helminth-infected vaccinated mice rapidly cleared the virus. In summary we demonstrate that basic improvements of influenza vaccination regimen are not sufficient to confer sterile immunity on the background of helminth-induced immunosuppression, despite amelioration of pathology i.e. weight loss. Our findings highlight the risk of failed vaccinations in helminth-endemic areas, especially in light of the ongoing vaccination campaign to control the COVID-19 pandemic.
Collapse
|
26
|
Mueller Brown K, Le Sage V, French AJ, Jones JE, Padovani GH, Avery AJ, Schultz-Cherry S, Rosch JW, Hiller NL, Lakdawala SS. Secondary infection with Streptococcus pneumoniae decreases influenza virus replication and is linked to severe disease. FEMS MICROBES 2022; 3:xtac007. [PMID: 35392116 PMCID: PMC8981988 DOI: 10.1093/femsmc/xtac007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/31/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022] Open
Abstract
Secondary bacterial infection is a common complication in severe influenza virus infections. During the H1N1 pandemic of 2009, increased mortality was observed among healthy young adults due to secondary bacterial pneumonia, one of the most frequent bacterial species being Streptococcus pneumoniae (Spn). Previous studies in mice and ferrets have suggested a synergistic relationship between Spn and influenza viruses. In this study, the ferret model was used to examine whether secondary Spn infection (strains BHN97 and D39) influence replication and airborne transmission of the 2009 pandemic H1N1 virus (H1N1pdm09). Secondary infection with Spn after H1N1pdm09 infection consistently resulted in a significant decrease in viral titers in the ferret nasal washes. While secondary Spn infection appeared to negatively impact influenza virus replication, animals precolonized with Spn were equally susceptible to H1N1pdm09 airborne transmission. In line with previous work, ferrets with preceding H1N1pdm09 and secondary Spn infection had increased bacterial loads and more severe clinical symptoms as compared to animals infected with H1N1pdm09 or Spn alone. Interestingly, the donor animals that displayed the most severe clinical symptoms had reduced airborne transmission of H1N1pdm09. Based on these data, we propose an asymmetrical relationship between these two pathogens, rather than a synergistic one, since secondary bacterial infection enhances Spn colonization and pathogenesis but decreases viral titers.
Collapse
Affiliation(s)
- Karina Mueller Brown
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Valerie Le Sage
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Bridgeside Point II, Pittsburgh, PA 15219, USA
| | - Andrea J French
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Bridgeside Point II, Pittsburgh, PA 15219, USA
| | - Jennifer E Jones
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Bridgeside Point II, Pittsburgh, PA 15219, USA
| | - Gabriella H Padovani
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Bridgeside Point II, Pittsburgh, PA 15219, USA
| | - Annika J Avery
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Bridgeside Point II, Pittsburgh, PA 15219, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Jason W Rosch
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - N Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Seema S Lakdawala
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Bridgeside Point II, Pittsburgh, PA 15219, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
Stetter N, Hartmann W, Brunn ML, Stanelle-Bertram S, Gabriel G, Breloer M. A Combination of Deworming and Prime-Boost Vaccination Regimen Restores Efficacy of Vaccination Against Influenza in Helminth-Infected Mice. Front Immunol 2022; 12:784141. [PMID: 34992602 PMCID: PMC8724120 DOI: 10.3389/fimmu.2021.784141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/03/2021] [Indexed: 11/17/2022] Open
Abstract
Helminths still infect a quarter of the human population. They manage to establish chronic infections by downmodulating the immune system of their hosts. Consequently, the immune response of helminth-infected individuals to vaccinations may be impaired as well. Here we study the impact of helminth-induced immunomodulation on vaccination efficacy in the mouse system. We have previously shown that an underlying Litomosoides sigmodontis infection reduced the antibody (Ab) response to anti-influenza vaccination in the context of a systemic expansion of type 1 regulatory T cells (Tr1). Most important, vaccine-induced protection from a challenge infection with the 2009 pandemic H1N1 influenza A virus (2009 pH1N1) was impaired in vaccinated, L. sigmodontis-infected mice. Here, we aim at the restoration of vaccination efficacy by drug-induced deworming. Treatment of mice with Flubendazole (FBZ) resulted in elimination of viable L. sigmodontis parasites in the thoracic cavity after two weeks. Simultaneous FBZ-treatment and vaccination did not restore Ab responses or protection in L. sigmodontis-infected mice. Likewise, FBZ-treatment two weeks prior to vaccination did not significantly elevate the influenza-specific Ig response and did not protect mice from a challenge infection with 2009 pH1N1. Analysis of the regulatory T cell compartment revealed that L. sigmodontis-infected and FBZ-treated mice still displayed expanded Tr1 cell populations that may contribute to the sustained suppression of vaccination responses in successfully dewormed mice. To outcompete this sustained immunomodulation in formerly helminth-infected mice, we finally combined the drug-induced deworming with an improved vaccination regimen. Two injections with the non-adjuvanted anti-influenza vaccine Begripal conferred 60% protection while MF59-adjuvanted Fluad conferred 100% protection from a 2009 pH1N1 infection in FBZ-treated, formerly L. sigmodontis-infected mice. Of note, applying this improved prime-boost regimen did not restore protection in untreated L. sigmodontis-infected mice. In summary our findings highlight the risk of failed vaccinations due to helminth infection.
Collapse
Affiliation(s)
- Nadine Stetter
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Wiebke Hartmann
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marie-Luise Brunn
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephanie Stanelle-Bertram
- Research Department for Viral Zoonoses - One Health, Leibniz Institute for Experimental Virology Heinrich Pette Institute (HPI), Hamburg, Germany
| | - Gülsah Gabriel
- Research Department for Viral Zoonoses - One Health, Leibniz Institute for Experimental Virology Heinrich Pette Institute (HPI), Hamburg, Germany.,Institute for Virology, University for Veterinary Medicine Hannover, Hannover, Germany
| | - Minka Breloer
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,Department for Biology, University Hamburg, Hamburg, Germany
| |
Collapse
|
28
|
Inherent heterogeneity of influenza A virus stability following aerosolization. Appl Environ Microbiol 2022; 88:e0227121. [PMID: 34985975 DOI: 10.1128/aem.02271-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Efficient human-to-human transmission represents a necessary adaptation for a zoonotic influenza A virus (IAV) to cause a pandemic. As such, many emerging IAVs are characterized for transmissibility phenotypes in mammalian models, with an emphasis on elucidating viral determinants of transmission and the role host immune responses contribute to mammalian adaptation. Investigations of virus infectivity and stability in aerosols concurrent with transmission assessments have increased in recent years, enhancing our understanding of this dynamic process. Here, we employ a diverse panel of 17 human and zoonotic IAVs, inclusive of seasonally circulating H1N1 and H3N2 viruses, and avian and swine viruses associated with human infection, to evaluate differences in spray factor (a value that assesses efficiency of the aerosolization process), stability, and infectivity following aerosolization. While most seasonal influenza viruses did not exhibit substantial variability within these parameters, there was more heterogeneity among zoonotic influenza viruses, which possess a diverse range of transmission phenotypes. Aging of aerosols at different relative humidities identified strain-specific levels of stability with different profiles identified between zoonotic H3, H5, and H7 subtype viruses associated with human infection. As studies continue to elucidate the complex components governing virus transmissibility, notably aerosol matrices and environmental parameters, considering the relative role of subtype- and strain-specific factors to modulate these parameters will improve our understanding of the pandemic potential of zoonotic influenza A viruses. Importance Transmission of respiratory pathogens through the air can facilitate the rapid and expansive spread of infection and disease through a susceptible population. While seasonal influenza viruses are quite capable of airborne spread, there is a lack of knowledge regarding how well influenza viruses remain viable after aerosolization, and if influenza viruses capable of jumping species barriers to cause human infection differ in this property from seasonal strains. We evaluated a diverse panel of influenza viruses associated with human infection (originating from human, avian, and swine reservoirs) for their ability to remain viable after aerosolization in the laboratory under a range of conditions. We found greater diversity among avian and swine-origin viruses compared with seasonal influenza viruses; strain-specific stability was also noted. Although influenza virus stability in aerosols is an underreported property, if molecular markers associated with enhanced stability are identified, we will be able to quickly recognize emerging strains of influenza that present the greatest pandemic threat.
Collapse
|
29
|
Gu Y, Zuo X, Zhang S, Ouyang Z, Jiang S, Wang F, Wang G. The Mechanism behind Influenza Virus Cytokine Storm. Viruses 2021; 13:1362. [PMID: 34372568 PMCID: PMC8310017 DOI: 10.3390/v13071362] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Influenza viruses are still a serious threat to human health. Cytokines are essential for cell-to-cell communication and viral clearance in the immune system, but excessive cytokines can cause serious immune pathology. Deaths caused by severe influenza are usually related to cytokine storms. The recent literature has described the mechanism behind the cytokine-storm network and how it can exacerbate host pathological damage. Biological factors such as sex, age, and obesity may cause biological differences between different individuals, which affects cytokine storms induced by the influenza virus. In this review, we summarize the mechanism behind influenza virus cytokine storms and the differences in cytokine storms of different ages and sexes, and in obesity.
Collapse
Affiliation(s)
| | | | | | | | | | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Y.G.); (X.Z.); (S.Z.); (Z.O.); (S.J.)
| | - Guoqiang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Y.G.); (X.Z.); (S.Z.); (Z.O.); (S.J.)
| |
Collapse
|
30
|
Daemi HB, Kulyar MFEA, He X, Li C, Karimpour M, Sun X, Zou Z, Jin M. Progression and Trends in Virus from Influenza A to COVID-19: An Overview of Recent Studies. Viruses 2021; 13:1145. [PMID: 34203647 PMCID: PMC8232279 DOI: 10.3390/v13061145] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/24/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Influenza is a highly known contagious viral infection that has been responsible for the death of many people in history with pandemics. These pandemics have been occurring every 10 to 30 years in the last century. The most recent global pandemic prior to COVID-19 was the 2009 influenza A (H1N1) pandemic. A decade ago, the H1N1 virus caused 12,500 deaths in just 19 months globally. Now, again, the world has been challenged with another pandemic. Since December 2019, the first case of a novel coronavirus (COVID-19) infection was detected in Wuhan. This infection has risen rapidly throughout the world; even the World Health Organization (WHO) announced COVID-19 as a worldwide emergency to ensure human health and public safety. This review article aims to discuss important issues relating to COVID-19, including clinical, epidemiological, and pathological features of COVID-19 and recent progress in diagnosis and treatment approaches for the COVID-19 infection. We also highlight key similarities and differences between COVID-19 and influenza A to ensure the theoretical and practical details of COVID-19.
Collapse
Affiliation(s)
- Hakimeh Baghaei Daemi
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (H.B.D.); (X.H.); (C.L.); (X.S.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan 430070, China
| | | | - Xinlin He
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (H.B.D.); (X.H.); (C.L.); (X.S.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan 430070, China
| | - Chengfei Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (H.B.D.); (X.H.); (C.L.); (X.S.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan 430070, China
| | - Morteza Karimpour
- Department of Biology, Azad University of Rasht, Rasht 4147654919, Iran;
| | - Xiaomei Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (H.B.D.); (X.H.); (C.L.); (X.S.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan 430070, China
| | - Zhong Zou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (H.B.D.); (X.H.); (C.L.); (X.S.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan 430070, China
| | - Meilin Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (H.B.D.); (X.H.); (C.L.); (X.S.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan 430070, China
| |
Collapse
|
31
|
Nguyen TQ, Rollon R, Choi YK. Animal Models for Influenza Research: Strengths and Weaknesses. Viruses 2021; 13:1011. [PMID: 34071367 PMCID: PMC8228315 DOI: 10.3390/v13061011] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
Influenza remains one of the most significant public health threats due to its ability to cause high morbidity and mortality worldwide. Although understanding of influenza viruses has greatly increased in recent years, shortcomings remain. Additionally, the continuous mutation of influenza viruses through genetic reassortment and selection of variants that escape host immune responses can render current influenza vaccines ineffective at controlling seasonal epidemics and potential pandemics. Thus, there is a knowledge gap in the understanding of influenza viruses and a corresponding need to develop novel universal vaccines and therapeutic treatments. Investigation of viral pathogenesis, transmission mechanisms, and efficacy of influenza vaccine candidates requires animal models that can recapitulate the disease. Furthermore, the choice of animal model for each research question is crucial in order for researchers to acquire a better knowledge of influenza viruses. Herein, we reviewed the advantages and limitations of each animal model-including mice, ferrets, guinea pigs, swine, felines, canines, and non-human primates-for elucidating influenza viral pathogenesis and transmission and for evaluating therapeutic agents and vaccine efficacy.
Collapse
Affiliation(s)
- Thi-Quyen Nguyen
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
| | - Rare Rollon
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
| | - Young-Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
- Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
32
|
Wang Z, Yu J, Sheng Z, Hause BM, Li F, Kaushik RS, Wang D. Functional study of a role of N-terminal HA stem region of swine influenza A virus in virus replication. Vet Microbiol 2021; 258:109132. [PMID: 34052744 DOI: 10.1016/j.vetmic.2021.109132] [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: 02/28/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022]
Abstract
Swine influenza A virus (SIV) is both a pathogen of economic significance to the swine industry and a potential zoonotic organism that may be transmitted to humans. We described here the detailed characterization of a role of N-terminal B-loop and CD helix of HA2 in swine influenza A virus replication. Results of our experiments demonstrated that Hemagglutinin (HA) protein of swine influenza virus could tolerate some mutations in functionally conserved B-loop and CD helix. These mutations, however, have substantially attenuated influenza virus replication in both cell lines and porcine primary tracheal epithelial cells. Significantly, we found that some B-loop or CD helix mutations generated virus mutants that replicated in MDCK and ST cell lines but failed to replicate in primary tracheal epithelial cells, thereby suggesting that swine HA protein may function as a viral virulence and pathogenesis factor. The described mutations may be further explored as attenuated vaccine candidates that can effectively prevent or eliminate the spread of influenza virus within and between swine herds.
Collapse
Affiliation(s)
- Zhao Wang
- China Institute of Veterinary Drug Control, 8 Zhongguancun South St, Beijing, China
| | - Jieshi Yu
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Zizhang Sheng
- Zuckerman Mind Brian Behavior Institute, Columbia University, New York, NY, USA
| | - Ben M Hause
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Feng Li
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Radhey S Kaushik
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA.
| | - Dan Wang
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
33
|
Xie C, Su W, Sia SF, Choy KT, Morrell S, Zhou J, Peiris M, Bloom J, Yen HL. A(H1N1)pdm09 influenza viruses replicating in ferret upper or lower respiratory tract differed in onward transmission potential by air. J Infect Dis 2021; 225:65-74. [PMID: 34036370 DOI: 10.1093/infdis/jiab286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/21/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A(H1N1)pdm09 influenza viruses replicate efficiently in respiratory epithelia and are transmitted via respiratory droplets and aerosols expelled by infected hosts. The relative onward transmission potential of influenza viruses replicating in the upper and lower respiratory epithelial cells has not been fully defined. METHODS Wild-type and barcoded A(H1N1)pdm09 viruses that differed by 2 synonymous mutations per gene segment were inoculated into ferrets via intra-nasal and intra-tracheal routes. Naïve recipients were exposed to the exhaled breath of inoculated donors for 8 hours on day 2 post-inoculation. Onward transmission potential of wild-type and barcoded genotypes were monitored by next generation sequencing. RESULTS Transmissible airborne particles were respired from the upper but not the lower respiratory epithelial cells of donor ferrets. There was limited mixing of viral populations replicating in the upper and lower respiratory tissues. CONCLUSIONS The ferret upper respiratory epithelium was mapped as the anatomic site that generated influenza virus-laden particles mediating onward transmission by air. Our results suggest that vaccines and antivirals should aim to reduce viral loads in the upper respiratory tract for prevention of influenza transmission.
Collapse
Affiliation(s)
- Chenyi Xie
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Wen Su
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Sin Fun Sia
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Ka-Tim Choy
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Steven Morrell
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Jie Zhou
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Jesse Bloom
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Howard Hughes Medical Institutes, Seattle, WA, USA
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
34
|
Saito M, Itoh Y, Yasui F, Munakata T, Yamane D, Ozawa M, Ito R, Katoh T, Ishigaki H, Nakayama M, Shichinohe S, Yamaji K, Yamamoto N, Ikejiri A, Honda T, Sanada T, Sakoda Y, Kida H, Le TQM, Kawaoka Y, Ogasawara K, Tsukiyama-Kohara K, Suga H, Kohara M. Macrocyclic peptides exhibit antiviral effects against influenza virus HA and prevent pneumonia in animal models. Nat Commun 2021; 12:2654. [PMID: 33976181 PMCID: PMC8113231 DOI: 10.1038/s41467-021-22964-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/06/2021] [Indexed: 12/20/2022] Open
Abstract
Most anti-influenza drugs currently used, such as oseltamivir and zanamivir, inhibit the enzymatic activity of neuraminidase. However, neuraminidase inhibitor-resistant viruses have already been identified from various influenza virus isolates. Here, we report the development of a class of macrocyclic peptides that bind the influenza viral envelope protein hemagglutinin, named iHA. Of 28 iHAs examined, iHA-24 and iHA-100 have inhibitory effects on the in vitro replication of a wide range of Group 1 influenza viruses. In particular, iHA-100 bifunctionally inhibits hemagglutinin-mediated adsorption and membrane fusion through binding to the stalk domain of hemagglutinin. Moreover, iHA-100 shows powerful efficacy in inhibiting the growth of highly pathogenic influenza viruses and preventing severe pneumonia at later stages of infection in mouse and non-human primate cynomolgus macaque models. This study shows the potential for developing cyclic peptides that can be produced more efficiently than antibodies and have multiple functions as next-generation, mid-sized biomolecules.
Collapse
Affiliation(s)
- Makoto Saito
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga, Japan
| | - Fumihiko Yasui
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Tsubasa Munakata
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Daisuke Yamane
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Makoto Ozawa
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Risa Ito
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takayuki Katoh
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hirohito Ishigaki
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga, Japan
| | - Misako Nakayama
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga, Japan
| | - Shintaro Shichinohe
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga, Japan
| | - Kenzaburo Yamaji
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Naoki Yamamoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Ai Ikejiri
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Tomoko Honda
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Takahiro Sanada
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Kida
- Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | | | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazumasa Ogasawara
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan.
| |
Collapse
|
35
|
Mettelman RC, Thomas PG. Human Susceptibility to Influenza Infection and Severe Disease. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a038711. [PMID: 31964647 PMCID: PMC8091954 DOI: 10.1101/cshperspect.a038711] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Influenza viruses are a persistent threat to global human health. Increased susceptibility to infection and the risk factors associated with progression to severe influenza-related disease are determined by a multitude of viral, host, and environmental conditions. Decades of epidemiologic research have broadly defined high-risk groups, while new genomic association studies have identified specific host factors impacting an individual's response to influenza. Here, we review and highlight both human susceptibility to influenza infection and the conditions that lead to severe influenza disease.
Collapse
Affiliation(s)
- Robert C Mettelman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
36
|
Abstract
Influenza infection poses significant risk for solid organ transplant recipients who often experience more severe infection with increased rates of complications, including those relating to the allograft. Although symptoms of influenza experienced by transplant recipients are similar to that of the general population, fever is not a ubiquitous symptom and lymphopenia is common. Annual inactivated influenza vaccine is recommended for all transplant recipients. Newer strategies such as using a higher dose vaccine or multiple doses in the same season appear to provide greater immunogenicity. Neuraminidase inhibitors are the mainstay of treatment and chemoprophylaxis although resistance may occur in the transplant setting. Influenza therapeutics are advancing, including the recent licensure of baloxavir; however, many remain to be evaluated in transplant recipients and are not yet in routine clinical use. Further population-based studies spanning multiple influenza seasons are needed to enhance our understanding of influenza epidemiology in solid organ transplant recipients. Specific assessment of newer influenza therapeutics in transplant recipients and refinement of prevention strategies are vital to reducing morbidity and mortality.
Collapse
Affiliation(s)
- Tina M Marinelli
- Division of Infectious Diseases, Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | | |
Collapse
|
37
|
Rioux M, Francis ME, Swan CL, Ge A, Kroeker A, Kelvin AA. The Intersection of Age and Influenza Severity: Utility of Ferrets for Dissecting the Age-Dependent Immune Responses and Relevance to Age-Specific Vaccine Development. Viruses 2021; 13:678. [PMID: 33920917 PMCID: PMC8071347 DOI: 10.3390/v13040678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
Many factors impact the host response to influenza virus infection and vaccination. Ferrets have been an indispensable reagent for influenza virus research for almost one hundred years. One of the most significant and well-known factors affecting human disease after infection is host age. Another significant factor is the virus, as strain-specific disease severity is well known. Studying age-related impacts on viral infection and vaccination outcomes requires an animal model that reflects both the physiological and immunological changes that occur with human aging, and sensitivity to differentially virulent influenza viruses. The ferret is uniquely susceptible to a plethora of influenza viruses impacting humans and has proven extremely useful in studying the clinical and immunological pictures of influenza virus infection. Moreover, ferrets developmentally have several of the age-related physiological changes that occur in humans throughout infancy, adulthood, old age, and pregnancy. In this review, we discuss ferret susceptibility to influenza viruses, summarize previous influenza studies using ferrets as models of age, and finally, highlight the application of ferret age models in the pursuit of prophylactic and therapeutic agents to address age-related influenza disease severity.
Collapse
Affiliation(s)
- Melissa Rioux
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H4R2, Canada; (M.R.); (A.G.)
| | - Magen E. Francis
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N5E3, Canada; (M.E.F.); (C.L.S.); (A.K.)
| | - Cynthia L. Swan
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N5E3, Canada; (M.E.F.); (C.L.S.); (A.K.)
| | - Anni Ge
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H4R2, Canada; (M.R.); (A.G.)
| | - Andrea Kroeker
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N5E3, Canada; (M.E.F.); (C.L.S.); (A.K.)
| | - Alyson A. Kelvin
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H4R2, Canada; (M.R.); (A.G.)
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N5E3, Canada; (M.E.F.); (C.L.S.); (A.K.)
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine, Dalhousie University, Halifax, NS B3K6R8, Canada
- The Canadian Center for Vaccinology (IWK Health Centre, Dalhousie University and the Nova Scotia Health Authority), Halifax, NS B3K6R8, Canada
- Department of Biochemistry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N5E5, Canada
| |
Collapse
|
38
|
Lv L, Gu S, Jiang H, Yan R, Chen Y, Chen Y, Luo R, Huang C, Lu H, Zheng B, Zhang H, Xia J, Tang L, Sheng G, Li L. Gut mycobiota alterations in patients with COVID-19 and H1N1 infections and their associations with clinical features. Commun Biol 2021; 4:480. [PMID: 33850296 PMCID: PMC8044104 DOI: 10.1038/s42003-021-02036-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/24/2021] [Indexed: 02/06/2023] Open
Abstract
The relationship between gut microbes and COVID-19 or H1N1 infections is not fully understood. Here, we compared the gut mycobiota of 67 COVID-19 patients, 35 H1N1-infected patients and 48 healthy controls (HCs) using internal transcribed spacer (ITS) 3-ITS4 sequencing and analysed their associations with clinical features and the bacterial microbiota. Compared to HCs, the fungal burden was higher. Fungal mycobiota dysbiosis in both COVID-19 and H1N1-infected patients was mainly characterized by the depletion of fungi such as Aspergillus and Penicillium, but several fungi, including Candida glabrata, were enriched in H1N1-infected patients. The gut mycobiota profiles in COVID-19 patients with mild and severe symptoms were similar. Hospitalization had no apparent additional effects. In COVID-19 patients, Mucoromycota was positively correlated with Fusicatenibacter, Aspergillus niger was positively correlated with diarrhoea, and Penicillium citrinum was negatively correlated with C-reactive protein (CRP). In H1N1-infected patients, Aspergillus penicilloides was positively correlated with Lachnospiraceae members, Aspergillus was positively correlated with CRP, and Mucoromycota was negatively correlated with procalcitonin. Therefore, gut mycobiota dysbiosis occurs in both COVID-19 patients and H1N1-infected patients and does not improve until the patients are discharged and no longer require medical attention. Lv et al. associate the gut mycobiota with clinical features and the bacterial microbiota by comparing COVID-19 patients to those infected with H1N1 and healthy controls. They find that gut mycobiota dysbiosis occurs in both COVID-19 patients and those infected with H1N1 and that it does not improve until patients no longer require medical attention, providing insights into a better healthcare guideline.
Collapse
Affiliation(s)
- Longxian Lv
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Silan Gu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huiyong Jiang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ren Yan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanfei Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yunbo Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Rui Luo
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chenjie Huang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Haifeng Lu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Beiwen Zheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hua Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lingling Tang
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital, Affiliated to Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Guoping Sheng
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital, Affiliated to Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
39
|
Le Sage V, Jones JE, Kormuth KA, Fitzsimmons WJ, Nturibi E, Padovani GH, Arevalo CP, French AJ, Avery AJ, Manivanh R, McGrady EE, Bhagwat AR, Lauring AS, Hensley SE, Lakdawala SS. Pre-existing heterosubtypic immunity provides a barrier to airborne transmission of influenza viruses. PLoS Pathog 2021; 17:e1009273. [PMID: 33600489 PMCID: PMC7891786 DOI: 10.1371/journal.ppat.1009273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/04/2021] [Indexed: 12/04/2022] Open
Abstract
Human-to-human transmission of influenza viruses is a serious public health threat, yet the precise role of immunity from previous infections on the susceptibility to airborne infection is still unknown. Using the ferret model, we examined the roles of exposure duration and heterosubtypic immunity on influenza transmission. We demonstrate that a 48 hour exposure is sufficient for efficient transmission of H1N1 and H3N2 viruses. To test pre-existing immunity, a gap of 8–12 weeks between primary and secondary infections was imposed to reduce innate responses and ensure robust infection of donor animals with heterosubtypic viruses. We found that pre-existing H3N2 immunity did not significantly block transmission of the 2009 H1N1pandemic (H1N1pdm09) virus to immune animals. Surprisingly, airborne transmission of seasonal H3N2 influenza strains was abrogated in recipient animals with H1N1pdm09 pre-existing immunity. This protection from natural infection with H3N2 virus was independent of neutralizing antibodies. Pre-existing immunity with influenza B virus did not block H3N2 virus transmission, indicating that the protection was likely driven by the adaptive immune response. We demonstrate that pre-existing immunity can impact susceptibility to heterologous influenza virus strains, and implicate a novel correlate of protection that can limit the spread of respiratory pathogens through the air. Influenza viruses pose a major public health threat through both seasonal epidemics and sporadic pandemics. An individual’s first influenza virus infection leaves long-lasting immunity, which plays an unknown role on susceptibility to airborne transmission of new viral strains. We show that pre-existing heterosubtypic immunity against the 2009 H1N1 pandemic virus protects recipient animals from airborne transmission of a seasonal H3N2 influenza virus, which is independent of cross-neutralizing antibodies. Pre-existing immunity with influenza B viruses was not protective suggesting that this phenomenon is driven by an adaptive response. Taken together, these data indicate that pre-existing immunity is an important barrier to airborne transmission and can influence the emergence and spread of potentially pandemic viruses.
Collapse
Affiliation(s)
- Valerie Le Sage
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jennifer E. Jones
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Karen A. Kormuth
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - William J. Fitzsimmons
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric Nturibi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Gabriella H. Padovani
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Claudia P. Arevalo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrea J. French
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Annika J. Avery
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Richard Manivanh
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth E. McGrady
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Amar R. Bhagwat
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Adam S. Lauring
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Seema S. Lakdawala
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
40
|
Seibert B, Angel M, Caceres CJ, Sutton T, Kumar A, Ferreri L, Cardenas-Garcia S, Geiger G, Rajao D, Perez DR. Development of a swine RNA polymerase I driven Influenza reverse genetics system for the rescue of type A and B Influenza viruses. J Virol Methods 2021; 288:114011. [PMID: 33152409 PMCID: PMC8103788 DOI: 10.1016/j.jviromet.2020.114011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 11/30/2022]
Abstract
Influenza viruses are among the most significant pathogens of humans and animals. Reverse genetics allows for the study of molecular attributes that modulate virus host range, virulence and transmission. The most common reverse genetics methods use bi-directional vectors containing a host RNA polymerase (pol) I promoter to produce virus-like RNAs and a host RNA pol II promoter to direct the synthesis of viral proteins. Given the species-dependency of the pol I promoter and virus-host interactions that influence replication of animal-origin influenza viruses in human-derived cells, we explored the potential of using the swine RNA pol I promoter (spol1) in a bi-directional vector for rescuing type A and B influenza viruses (IAV and IBV, respectively) in swine and human cells. The spol1-based bi-directional plasmid vector led to efficient rescue of IAVs of different origins (human, swine, and avian) as well as IBV in both swine- and human-origin tissue culture cells. In addition, virus rescue was successful using a recombinant bacmid containing all eight segments of a swine origin IAV. In conclusion, the spol1-based reverse genetics system is a new platform to study influenza viruses and produce swine influenza vaccines with increased transfection efficiency.
Collapse
Affiliation(s)
- Brittany Seibert
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Matthew Angel
- Cellular Biology Section, Laboratory of Viral Diseases NIAID, NIH, Bethesda, MD, United States
| | - C Joaquin Caceres
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Troy Sutton
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, United States
| | - Ayush Kumar
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Lucas Ferreri
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Stivalis Cardenas-Garcia
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Ginger Geiger
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Daniela Rajao
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States
| | - Daniel R Perez
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, GA, United States.
| |
Collapse
|
41
|
Raynor PC, Adesina A, Aboubakr HA, Yang M, Torremorell M, Goyal SM. Comparison of samplers collecting airborne influenza viruses: 1. Primarily impingers and cyclones. PLoS One 2021; 16:e0244977. [PMID: 33507951 PMCID: PMC7842955 DOI: 10.1371/journal.pone.0244977] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022] Open
Abstract
Researchers must be able to measure concentrations, sizes, and infectivity of virus-containing particles in animal agriculture facilities to know how far infectious virus-containing particles may travel through air, where they may deposit in the human or animal respiratory tract, and the most effective ways to limit exposures to them. The objective of this study was to evaluate a variety of impinger and cyclone aerosol or bioaerosol samplers to determine approaches most suitable for detecting and measuring concentrations of virus-containing particles in air. Six impinger/cyclone air samplers, a filter-based sampler, and a cascade impactor were used in separate tests to collect artificially generated aerosols of MS2 bacteriophage and swine and avian influenza viruses. Quantification of infectious MS2 coliphage was carried out using a double agar layer procedure. The influenza viruses were titrated in cell cultures to determine quantities of infectious virus. Viral RNA was extracted and used for quantitative real time RT-PCR, to provide total virus concentrations for all three viruses. The amounts of virus recovered and the measured airborne virus concentrations were calculated and compared among the samplers. Not surprisingly, high flow rate samplers generally collected greater quantities of virus than low flow samplers. However, low flow rate samplers generally measured higher, and likely more accurate, airborne concentrations of Infectious virus and viral RNA than high flow samplers. To assess airborne viruses in the field, a two-sampler approach may work well. A suitable high flow sampler may provide low limits of detection to determine if any virus is present in the air. If virus is detected, a suitable lower flow sampler may measure airborne virus concentrations accurately.
Collapse
Affiliation(s)
- Peter C. Raynor
- Division of Environmental Health Sciences, University of Minnesota, School of Public Health, Minneapolis, Minnesota, United States of America
| | - Adepeju Adesina
- Division of Environmental Health Sciences, University of Minnesota, School of Public Health, Minneapolis, Minnesota, United States of America
| | - Hamada A. Aboubakr
- University of Minnesota, College of Veterinary Medicine, Veterinary Population Medicine Department, St. Paul, Minnesota, United States of America
| | - My Yang
- University of Minnesota, College of Veterinary Medicine, Veterinary Population Medicine Department, St. Paul, Minnesota, United States of America
| | - Montserrat Torremorell
- University of Minnesota, College of Veterinary Medicine, Veterinary Population Medicine Department, St. Paul, Minnesota, United States of America
| | - Sagar M. Goyal
- University of Minnesota, College of Veterinary Medicine, Veterinary Population Medicine Department, St. Paul, Minnesota, United States of America
| |
Collapse
|
42
|
Dose-dependent response to infection with SARS-CoV-2 in the ferret model and evidence of protective immunity. Nat Commun 2021; 12:81. [PMID: 33398055 PMCID: PMC7782478 DOI: 10.1038/s41467-020-20439-y] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022] Open
Abstract
There is a vital need for authentic COVID-19 animal models to enable the pre-clinical evaluation of candidate vaccines and therapeutics. Here we report a dose titration study of SARS-CoV-2 in the ferret model. After a high (5 × 106 pfu) and medium (5 × 104 pfu) dose of virus is delivered, intranasally, viral RNA shedding in the upper respiratory tract (URT) is observed in 6/6 animals, however, only 1/6 ferrets show similar signs after low dose (5 × 102 pfu) challenge. Following sequential culls pathological signs of mild multifocal bronchopneumonia in approximately 5–15% of the lung is seen on day 3, in high and medium dosed groups. Ferrets re-challenged, after virus shedding ceased, are fully protected from acute lung pathology. The endpoints of URT viral RNA replication & distinct lung pathology are observed most consistently in the high dose group. This ferret model of SARS-CoV-2 infection presents a mild clinical disease. SARS-CoV-2 induces mild infection in ferret model. Here, Ryan et al. characterise optimal infection dosage inducing upper respiratory tract (UTR) viral shedding, progression time of viral shedding, and pathology in ferrets and finally provide evidence for protection after re-challenge.
Collapse
|
43
|
Hartmann W, Brunn ML, Stetter N, Gagliani N, Muscate F, Stanelle-Bertram S, Gabriel G, Breloer M. Helminth Infections Suppress the Efficacy of Vaccination against Seasonal Influenza. Cell Rep 2020; 29:2243-2256.e4. [PMID: 31747598 DOI: 10.1016/j.celrep.2019.10.051] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/29/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Helminth parasites infect more than a quarter of the human population and inflict significant changes to the immunological status of their hosts. Here, we analyze the impact of helminth infections on the efficacy of vaccinations using Litomosoides sigmodontis-infected mice. Concurrent helminth infection reduces the quantity and quality of antibody responses to vaccination against seasonal influenza. Vaccination-induced protection against challenge infections with the human pathogenic 2009 pandemic H1N1 influenza A virus is drastically impaired in helminth-infected mice. Impaired responses are also observed if vaccinations are performed after clearance of a previous helminth infection, suggesting that individuals in helminth-endemic areas may not always benefit from vaccinations, even in the absence of an acute and diagnosable helminth infection. Mechanistically, the suppression is associated with a systemic and sustained expansion of interleukin (IL)-10-producing CD4+CD49+LAG-3+ type 1 regulatory T cells and partially abrogated by in vivo blockade of the IL-10 receptor.
Collapse
Affiliation(s)
- Wiebke Hartmann
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Marie-Luise Brunn
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Nadine Stetter
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Nicola Gagliani
- I Department of Medicine and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, 17176 Stockholm, Sweden
| | - Franziska Muscate
- I Department of Medicine and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stephanie Stanelle-Bertram
- Research Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Gülsah Gabriel
- Research Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany; Institute for Virology, University for Veterinary Medicine, Hannover, Germany
| | - Minka Breloer
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany.
| |
Collapse
|
44
|
Pawar S, Hsu L, Narendar Reddy T, Ravinder M, Ren CT, Lin YW, Cheng YY, Lin TW, Hsu TL, Wang SK, Wong CH, Wu CY. Synthesis of Asymmetric N-Glycans as Common Core Substrates for Structural Diversification through Selective Enzymatic Glycosylation. ACS Chem Biol 2020; 15:2382-2394. [PMID: 32830946 DOI: 10.1021/acschembio.0c00359] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
N-glycans on the cell surface provide distinct signatures that are recognized by different glycan-binding proteins (GBPs) and pathogens. Most glycans in humans are asymmetric and isomeric, yet their biological functions are not well understood due to their lack of availability for studies. In this work, we have developed an improved strategy for asymmetric N-glycan assembly and diversification using designed common core substrates prepared chemically for selective enzymatic fucosylation and sialylation. The resulting 26 well-defined glycans that carry the sialic acid residue on different antennae were used in a microarray as a representative application to profile the binding specificity of hemagglutinin (HA) from the avian influenza virus (H5N2). We found distinct binding affinity for the Neu5Ac-Gal epitope linked to the N-acetylglucosamine (GlcNAc) of different branches and only a minor effect in binding for the terminal galactose on different branches. Overall, the microarray analysis showed branch-biased and context-based recognition patterns.
Collapse
Affiliation(s)
- Sujeet Pawar
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, Taiwan
| | - Li Hsu
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
- Department of Chemistry, National Taiwan University, Taipei, 106 Taiwan
| | - Thatikonda Narendar Reddy
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Mettu Ravinder
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Chien-Tai Ren
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Yu-Wei Lin
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Yang-Yu Cheng
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Tzu-Wen Lin
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Tsui-Ling Hsu
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
| | - Sheng-Kai Wang
- Department of Chemistry, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, Taiwan
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529 Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
45
|
Genetically and Antigenically Divergent Influenza A(H9N2) Viruses Exhibit Differential Replication and Transmission Phenotypes in Mammalian Models. J Virol 2020; 94:JVI.00451-20. [PMID: 32611751 DOI: 10.1128/jvi.00451-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/20/2020] [Indexed: 12/15/2022] Open
Abstract
Low-pathogenicity avian influenza A(H9N2) viruses, enzootic in poultry populations in Asia, are associated with fewer confirmed human infections but higher rates of seropositivity compared to A(H5) or A(H7) subtype viruses. Cocirculation of A(H5) and A(H7) viruses leads to the generation of reassortant viruses bearing A(H9N2) internal genes with markers of mammalian adaptation, warranting continued surveillance in both avian and human populations. Here, we describe active surveillance efforts in live poultry markets in Vietnam in 2018 and compare representative viruses to G1 and Y280 lineage viruses that have infected humans. Receptor binding properties, pH thresholds for HA activation, in vitro replication in human respiratory tract cells, and in vivo mammalian pathogenicity and transmissibility were investigated. While A(H9N2) viruses from both poultry and humans exhibited features associated with mammalian adaptation, one human isolate from 2018, A/Anhui-Lujiang/39/2018, exhibited increased capacity for replication and transmission, demonstrating the pandemic potential of A(H9N2) viruses.IMPORTANCE A(H9N2) influenza viruses are widespread in poultry in many parts of the world and for over 20 years have sporadically jumped species barriers to cause human infection. As these viruses continue to diversify genetically and antigenically, it is critical to closely monitor viruses responsible for human infections, to ascertain if A(H9N2) viruses are acquiring properties that make them better suited to infect and spread among humans. In this study, we describe an active poultry surveillance system established in Vietnam to identify the scope of influenza viruses present in live bird markets and the threat they pose to human health. Assessment of a recent A(H9N2) virus isolated from an individual in China in 2018 is also reported, and it was found to exhibit properties of adaptation to humans and, importantly, it shows similarities to strains isolated from the live bird markets of Vietnam.
Collapse
|
46
|
Characterization of highly pathogenic avian influenza H5Nx viruses in the ferret model. Sci Rep 2020; 10:12700. [PMID: 32728042 PMCID: PMC7391700 DOI: 10.1038/s41598-020-69535-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/08/2020] [Indexed: 01/06/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5 viruses, of the A/goose/Guangdong/1/1996 lineage, have exhibited substantial geographic spread worldwide since the first detection of H5N1 virus in 1996. Accumulation of mutations in the HA gene has resulted in several phylogenetic clades, while reassortment with other avian influenza viruses has led to the emergence of new virus subtypes (H5Nx), notably H5N2, H5N6, and H5N8. H5Nx viruses represent a threat to both the poultry industry and human health and can cause lethal human disease following virus exposure. Here, HPAI H5N6 and H5N2 viruses (isolated between 2014 and 2017) of the 2.3.4.4 clade were assessed for their capacity to replicate in human respiratory tract cells, and to cause disease and transmit in the ferret model. All H5N6 viruses possessed increased virulence in ferrets compared to the H5N2 virus; however, pathogenicity profiles varied among the H5N6 viruses tested, from mild infection with sporadic virus dissemination beyond the respiratory tract, to severe disease with fatal outcome. Limited transmission between co-housed ferrets was observed with the H5N6 viruses but not with the H5N2 virus. In vitro evaluation of H5Nx virus replication in Calu-3 cells and the identification of mammalian adaptation markers in key genes associated with pathogenesis supports these findings.
Collapse
|
47
|
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
|
48
|
Vo TS, Vo TTTN, Vo TTBC. Coronavirus Infection Prevention by Wearing Masks. Eurasian J Med 2020; 52:197-201. [PMID: 32612431 PMCID: PMC7311128 DOI: 10.5152/eurasianjmed.2020.20056] [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: 03/24/2020] [Accepted: 04/09/2020] [Indexed: 11/22/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) [2019-nCoV; severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] was first detected in Wuhan, China at the end of 2019. In current status, spread of COVID-19 in person-to-person could be caused mainly by respiratory droplets, which leads to the spread of the influenza virus in both community and clinicians. Thus, in order to reduce the risk of that, the urgent management strategies against COVID-19 are to block transmission, isolation, protection, and using drug or vaccine updated on an ongoing basis. unfortunately, no drugs or vaccines still has yet been allowed to treat patients with COVID-19, so the rapid detection of effective intercessions against COVID-19 is seemed a major challenge on the all world. Herein, this article attempts summarizing to introduce the characterization of COVID-19, the influence of droplets travel in person-to-person transmission and the effect of wearing masks in the infection prevention of influenza virus, as well as understanding its advantage and role in the coronavirus infection prevention.
Collapse
Affiliation(s)
- Thi Sinh Vo
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tran Thi Thu Ngoc Vo
- Department of Acupuntur, Tuina and Moxibustion, Anhui University of Traditional Chinese Medicine, Anhui, China
- Qui Nhon City Hospital, Binh Dinh, Vietnam
| | | |
Collapse
|
49
|
Avian Influenza A Virus Infects Swine Airway Epithelial Cells without Prior Adaptation. Viruses 2020; 12:v12060589. [PMID: 32481674 PMCID: PMC7374723 DOI: 10.3390/v12060589] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/15/2023] Open
Abstract
Pigs play an important role in the interspecies transmission of influenza A viruses (IAV). The porcine airway epithelium contains binding sites for both swine/human IAV (α2,6-linked sialic acids) and avian IAV (α2,3-linked sialic acids) and therefore is suited for adaptation of viruses from other species as suggested by the “mixing vessel theory”. Here, we applied well-differentiated swine airway epithelial cells to find out whether efficient infection by avian IAV requires prior adaption. Furthermore, we analyzed the influence of the sialic acid-binding activity and the virus-induced detrimental effects. Surprisingly, an avian IAV H1N1 strain circulating in European poultry and waterfowl shows increased and prolonged viral replication without inducing a strong innate immune response. This virus could infect the lower respiratory tract in our precision cut-lung slice model. Pretreating the cells with poly (I:C) and/or JAK/STAT pathway inhibitors revealed that the interferon-stimulated innate immune response influences the replication of avian IAV in swine airway epitheliums but not that of swine IAV. Further studies indicated that in the infection by IAVs, the binding affinity of sialic acid is not the sole factor affecting the virus infectivity for swine or human airway epithelial cells, whereas it may be crucial in well-differentiated ferret tracheal epithelial cells. Taken together, our results suggest that the role of pigs being the vessel of interspecies transmission should be reconsidered, and the potential of avian H1N1 viruses to infect mammals needs to be characterized in more detail.
Collapse
|
50
|
Park MS, Kim JI, Bae JY, Park MS. Animal models for the risk assessment of viral pandemic potential. Lab Anim Res 2020; 36:11. [PMID: 32337177 PMCID: PMC7175453 DOI: 10.1186/s42826-020-00040-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Pandemics affect human lives severely and globally. Experience predicts that there will be a pandemic for sure although the time is unknown. When a viral epidemic breaks out, assessing its pandemic risk is an important part of the process that characterizes genomic property, viral pathogenicity, transmission in animal model, and so forth. In this review, we intend to figure out how a pandemic may occur by looking into the past influenza pandemic events. We discuss interpretations of the experimental evidences resulted from animal model studies and extend implications of viral pandemic potentials and ingredients to emerging viral epidemics. Focusing on the pandemic potential of viral infectious diseases, we suggest what should be assessed to prevent global catastrophes from influenza virus, Middle East respiratory syndrome coronavirus, dengue and Zika viruses.
Collapse
Affiliation(s)
- Mee Sook Park
- Department of Microbiology, Institute for Viral Diseases, College of Medicine, Korea University, Seoul, Republic of Korea 02841
| | - Jin Il Kim
- Department of Microbiology, Institute for Viral Diseases, College of Medicine, Korea University, Seoul, Republic of Korea 02841
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, College of Medicine, Korea University, Seoul, Republic of Korea 02841
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, College of Medicine, Korea University, Seoul, Republic of Korea 02841
| |
Collapse
|