1
|
Salve BG, Sharma S, Vijay N. Evolutionary diversity of CXCL16-CXCR6: Convergent substitutions and recurrent gene loss in sauropsids. Immunogenetics 2024; 76:397-415. [PMID: 39400711 DOI: 10.1007/s00251-024-01357-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
The CXCL16-CXCR6 axis is crucial for regulating the persistence of CD8 tissue-resident memory T cells (TRM). CXCR6 deficiency lowers TRM cell numbers in the lungs and depletes ILC3s in the lamina propria, impairing mucosal defence. This axis is linked to diseases like HIV/SIV, cancer, and COVID-19. Together, these highlight that the CXCL16-CXCR6 axis is pivotal in host immunity. Previous studies of the CXCL16-CXCR6 axis found genetic variation among species but were limited to primates and rodents. To understand the evolution and diversity of CXCL16-CXCR6 across vertebrates, we compared approximately 400 1-to-1 CXCR6 orthologs spanning diverse vertebrates. The unique DRF motif of CXCR6 facilitates leukocyte adhesion by interacting with cell surface-expressed CXCL16 and plays a key role in G-protein selectivity during receptor signalling; however, our findings show that this motif is not universal. The DRF motif is restricted to mammals, turtles, and frogs, while the DRY motif, typical in other CKRs, is found in snakes and lizards. Most birds exhibit the DRL motif. These substitutions at the DRF motif affect the receptor-Gi/o protein interaction. We establish recurrent CXCR6 gene loss in 10 out of 36 bird orders, including Galliformes and Passeriformes, Crocodilia, and Elapidae, attributed to segmental deletions and/or frame-disrupting changes. Notably, single-cell RNA sequencing of the lung shows a drop in TRM cells in species with CXCR6 loss, suggesting a possible link. The concurrent loss of ITGAE, CXCL16, and CXCR6 in chickens may have altered CD8 TRM cell abundance, with implications for immunity against viral diseases and vaccines inducing CD8 TRM cells.
Collapse
Affiliation(s)
- Buddhabhushan Girish Salve
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India
| | - Sandhya Sharma
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India
| | - Nagarjun Vijay
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India.
| |
Collapse
|
2
|
Marchenko VY, Panova AS, Kolosova NP, Gudymo AS, Svyatchenko SV, Danilenko AV, Vasiltsova NN, Egorova ML, Onkhonova GS, Zhestkov PD, Zinyakov NG, Andreychuk DB, Chvala IA, Kosenko MN, Moiseeva AA, Boldyrev ND, Shadrinova KN, Perfilieva ON, Ryzhikov AB. Characterization of H5N1 avian influenza virus isolated from bird in Russia with the E627K mutation in the PB2 protein. Sci Rep 2024; 14:26490. [PMID: 39489822 DOI: 10.1038/s41598-024-78175-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
Currently A(H5Nx) avian influenza viruses are globally widespread and continue to evolve. Since their emergence in 2020 novel highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b reassortant viruses have become predominant in the world and caused multiple infections in mammals. It was shown that some of A(H5N1) viruses mostly isolated from mammals contain an E627K mutation in the PB2 protein which can lead to adaptation of influenza viruses to mammalian cells. In 2023 in Russia we have isolated two highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b viruses from birds one of which contained an E627K mutation in the PB2 protein. This virus had increased virulence in mice. Limited airborne transmission of the virus with the PB2-E627K mutation was observed between ferrets, in which infectious virus was detected in the nasal washings of the three of the twelve recipient ferrets, and clinical symptoms of the disease were observed in one case. Both viruses showed dominant binding to avian-type sialoside receptors, which was most likely the reason for the limited transmissibility. Thus, this study indicates a possible limited increase in the pandemic potential of A(H5N1) 2.3.4.4b viruses and highlights the importance of continuous avian influenza surveillance for pandemic preparedness and response.
Collapse
Affiliation(s)
- Vasiliy Yu Marchenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia.
| | - Anastasia S Panova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Natalia P Kolosova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Andrey S Gudymo
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Svetlana V Svyatchenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Alexey V Danilenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Natalia N Vasiltsova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Marina L Egorova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Galina S Onkhonova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Pavel D Zhestkov
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Nikolay G Zinyakov
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Dmitriy B Andreychuk
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Ilya A Chvala
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Maksim N Kosenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Anastasia A Moiseeva
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Nikita D Boldyrev
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Kiunnei N Shadrinova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Olga N Perfilieva
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Alexander B Ryzhikov
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| |
Collapse
|
3
|
Elhusseiny MH, Elsayed MM, Mady WH, Mahana O, Bakry NR, Abdelaziz O, Arafa AS, Shahein MA, Eid S, Naguib MM. Genetic features of avian influenza (A/H5N8) clade 2.3.4.4b isolated from quail in Egypt. Virus Res 2024; 350:199482. [PMID: 39396573 DOI: 10.1016/j.virusres.2024.199482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Several genotypes of the highly pathogenic avian influenza (HPAI) virus H5N8 subtype within clade 2.3.4.4b continue to circulate in different species of domestic birds across Egypt. It is believed that quail contribute to virus replication and adaptation to other gallinaceous poultry species and humans. This study provides genetic characterization of the full genome of HPAI H5N8 isolated from quail in Egypt. The virus was isolated from a commercial quail farm associated with respiratory signs. To characterize the genetic features of the detected virus, gene sequencing via Sanger technology and phylogenetic analysis were performed. The results revealed high nucleotide identity with the HPAI H5N8 virus from Egypt, which has multiple basic amino acid motifs PLREKRRKR/GLF at the hemagglutinin (HA) cleavage site. Phylogenetic analysis of the eight gene segments revealed that the quail isolate is grouped with HPAI H5N8 viruses of clade 2.3.4.4b and closely related to the most recent circulating H5N8 viruses in Egypt. Whole-genome characterization revealed amino acid preferences for avian receptors with few mutations, indicating their affinity for human-like receptors and increased virulence in mammals, such as S123P, S133A, T156A and A263T in the HA gene. In addition, the sequencing results revealed a lack of markers associated with influenza antiviral resistance in the neuraminidase and matrix-2 coding proteins. The results of the present study support the spread of HPAIV H5N8 to species other than chickens in Egypt. Therefore, continuous surveillance of AIV in different bird species in Egypt followed by full genomic characterization is needed for better virus control and prevention.
Collapse
Affiliation(s)
- Mohamed H Elhusseiny
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Moataz M Elsayed
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Wesam H Mady
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Osama Mahana
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Neveen R Bakry
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Ola Abdelaziz
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Abdel-Sattar Arafa
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | | | - Samah Eid
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Mahmoud M Naguib
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt; Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK.
| |
Collapse
|
4
|
Ohlopkova OV, Goncharov AE, Aslanov BI, Fadeev AV, Davidyuk YN, Moshkin AD, Stolbunova KA, Stepanyuk MA, Sobolev IA, Tyumentseva MA, Tyumentsev AI, Shestopalov AM, Akimkin VG. First detection of influenza A virus subtypes H1N1 and H3N8 in the Antarctic region: King George Island, 2023. Vopr Virusol 2024; 69:377-389. [PMID: 39361931 DOI: 10.36233/0507-4088-257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Indexed: 10/05/2024]
Abstract
RELEVANCE Influenza A virus is characterized by a segmented single-stranded RNA genome. Such organization of the virus genome determines the possibility of reassortment, which can lead to the emergence of new virus variants. The main natural reservoir of most influenza A virus subtypes are wild waterfowl. Seasonal migrations gather waterfowl from all major migration routes to nesting areas near the northern and southern polar circles. This makes intercontinental spread of influenza A viruses possible. Objective ‒ to conduct molecular genetic monitoring and study the phylogenetic relationships of influenza A virus variants circulating in Antarctica in 2023. MATERIALS AND METHODS We studied 84 samples of biological material obtained from birds and marine mammals in April‒May 2023 in coastal areas of Antarctica. For 3 samples, sequencing was performed on the Miseq, Illumina platform and phylogenetic analysis of the obtained nucleotide sequences of the influenza A virus genomes was performed. RESULTS The circulation of avian influenza virus in the Antarctic region was confirmed. Heterogeneity of the pool of circulating variants of the influenza A virus (H3N8, H1N1) was revealed. Full-length genomes of the avian influenza virus were sequenced and posted in the GISAID database (EPI_ISL_19032103, 19174530, 19174467). CONCLUSION The study of the genetic diversity of influenza A viruses circulating in the polar regions of the Earth and the identification of the conditions for the emergence of new genetic variants is a relevant task for the development of measures to prevent biological threats.
Collapse
Affiliation(s)
- O V Ohlopkova
- Central Research Institute of Epidemiology, Rospotrebnadzor
- Federal Research Center for Fundamental and Translational Medicine
| | - A E Goncharov
- Institute of Experimental Medicine
- Northwestern State Medical University named after I.I. Mechnikov
| | - B I Aslanov
- Northwestern State Medical University named after I.I. Mechnikov
| | - A V Fadeev
- A.A. Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation
| | - Y N Davidyuk
- Federal State Educational Institution of Higher Education «Kazan (Volga Region) Federal University»
| | - A D Moshkin
- Federal Research Center for Fundamental and Translational Medicine
| | - K A Stolbunova
- Federal Research Center for Fundamental and Translational Medicine
| | - M A Stepanyuk
- Federal Research Center for Fundamental and Translational Medicine
| | - I A Sobolev
- Federal Research Center for Fundamental and Translational Medicine
| | | | - A I Tyumentsev
- Central Research Institute of Epidemiology, Rospotrebnadzor
| | - A M Shestopalov
- Federal Research Center for Fundamental and Translational Medicine
| | - V G Akimkin
- Central Research Institute of Epidemiology, Rospotrebnadzor
| |
Collapse
|
5
|
Goecker ZC, Burke MC, Remoroza CA, Liu Y, Mirokhin YA, Sheetlin SL, Tchekhovskoi DV, Yang X, Stein SE. Variation of Site-Specific Glycosylation Profiles of Recombinant Influenza Glycoproteins. Mol Cell Proteomics 2024; 23:100827. [PMID: 39128790 PMCID: PMC11417209 DOI: 10.1016/j.mcpro.2024.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/08/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024] Open
Abstract
This work presents a detailed determination of site-specific N-glycan distributions of the recombinant influenza glycoproteins hemagglutinin (HA) and neuraminidase. Variation in glycosylation among recombinant glycoproteins is not predictable and can depend on details of the biomanufacturing process as well as details of protein structure. In this study, recombinant influenza proteins were analyzed from eight strains of four different suppliers. These include five HA and three neuraminidase proteins, each produced from a HEK293 cell line. Digestion was conducted using a series of complex multienzymatic methods designed to isolate glycopeptides containing single N-glycosylated sites. Site-specific glycosylation profiles of intact glycopeptides were produced using a recently developed method and comparisons were made using spectral similarity scores. Variation in glycan abundances and distribution was most pronounced between different strains of virus (similarity score = 383 out of 999), whereas digestion replicates and injection replicates showed relatively little variation (similarity score = 957). Notably, glycan distributions for homologous regions of influenza glycoprotein variants showed low variability. Due to the multiple possible sources of variation and inherent analytical difficulties in site-specific glycan determinations, variations were individually examined for multiple factors, including differences in supplier, production batch, protease digestion, and replicate measurement. After comparing all glycosylation distributions, four distinguishable classes could be identified for the majority of sites. Finally, attempts to identify glycosylation distributions on adjacent potential N-glycosylated sites of one HA variant were made. Only the second site (NnST) was found to be occupied using two rarely used proteases in proteomics, subtilisin and esperase, both of which did selectively cleave these adjacent sites.
Collapse
Affiliation(s)
- Zachary C Goecker
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA.
| | - Meghan C Burke
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Concepcion A Remoroza
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yi Liu
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yuri A Mirokhin
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Sergey L Sheetlin
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Dmitrii V Tchekhovskoi
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Xiaoyu Yang
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Stephen E Stein
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
6
|
Rajanala K, Upadhyay AK. Vaccines for Respiratory Viruses-COVID and Beyond. Vaccines (Basel) 2024; 12:936. [PMID: 39204059 PMCID: PMC11360283 DOI: 10.3390/vaccines12080936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 (coronavirus disease 2019) pandemic had an extensive impact on global morbidity and mortality. Several other common respiratory viruses, such as the influenza virus and respiratory syncytial virus (RSV), are endemic or epidemic agents causing acute respiratory infections that are easily transmissible and pose a significant threat to communities due to efficient person-to-person transmission. These viruses can undergo antigenic variation through genetic mutations, resulting in the emergence of novel strains or variants, thereby diminishing the effectiveness of current vaccines, and necessitating ongoing monitoring and adjustment of vaccine antigens. As the virus-specific immunity is maintained only for several weeks or months after the infection, there is an emergent need to develop effective and durable vaccines. Additionally, specific populations, such as elderly or immunocompromised individuals, may exhibit reduced immune responses to respiratory viruses, posing significant challenges to develop vaccines that elicit durable and potent immunity. We present a comprehensive review of the molecular mechanisms underlying the pathogenesis and virulence of common respiratory viruses, such as RSV, influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We discuss several vaccine approaches that are under development. A thorough understanding of the current strategies and the challenges encountered during the vaccine development process can lead to the advancement of effective next-generation vaccines.
Collapse
|
7
|
Vaghela S, Welch VL, Sinh A, Di Fusco M. Caregiver Burden among Patients with Influenza or Influenza-like Illness (ILI): A Systematic Literature Review. Healthcare (Basel) 2024; 12:1591. [PMID: 39201150 PMCID: PMC11353737 DOI: 10.3390/healthcare12161591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/27/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Influenza and influenza-like illness (ILI) pose significant clinical and economic burdens globally each year. This systematic literature review examined quantitative studies evaluating the impact of patients' influenza/ILI on their caregivers' well-being, focusing on health-related quality of life (HRQoL), work productivity, and activity impairment. A comprehensive search across six databases, including the Cochrane Database of Systematic Reviews, Embase, MEDLINE via PubMed, Ovid, PsycNet, and Web of Science, yielded 18,689 records, of which 13,156 abstracts were screened, and 662 full-text articles were reviewed from January 2007 to April 2024. Thirty-six studies [HRQoL: 2; productivity: 33; both: 1] covering 22 countries were included. Caregivers of 47,758 influenza or ILI patients across 123 study cohorts were assessed in the review. The mean workday loss among caregivers ranged from 0.5 to 10.7 days per episode, influenced by patients' influenza status (positive or negative), disease severity (mild or moderate-to-severe), age, viral type (influenza A or B), and vaccination/treatment usage. The HRQoL of caregivers, including their physical and emotional well-being, was affected by a patient's influenza or ILI, where the severity and duration of a patient's illness were associated with worse HRQoL. This review shows that the consequences of influenza or ILI significantly affect not only patients but also their caregivers.
Collapse
Affiliation(s)
- Shailja Vaghela
- HealthEcon Consulting, Inc., Ancaster, ON L9G 4L2, Canada; (S.V.)
| | | | - Anup Sinh
- HealthEcon Consulting, Inc., Ancaster, ON L9G 4L2, Canada; (S.V.)
| | | |
Collapse
|
8
|
Karakus U, Mena I, Kottur J, El Zahed SS, Seoane R, Yildiz S, Chen L, Plancarte M, Lindsay L, Halpin R, Stockwell TB, Wentworth DE, Boons GJ, Krammer F, Stertz S, Boyce W, de Vries RP, Aggarwal AK, García-Sastre A. H19 influenza A virus exhibits species-specific MHC class II receptor usage. Cell Host Microbe 2024; 32:1089-1102.e10. [PMID: 38889725 PMCID: PMC11295516 DOI: 10.1016/j.chom.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/01/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024]
Abstract
Avian influenza A virus (IAV) surveillance in Northern California, USA, revealed unique IAV hemagglutinin (HA) genome sequences in cloacal swabs from lesser scaups. We found two closely related HA sequences in the same duck species in 2010 and 2013. Phylogenetic analyses suggest that both sequences belong to the recently discovered H19 subtype, which thus far has remained uncharacterized. We demonstrate that H19 does not bind the canonical IAV receptor sialic acid (Sia). Instead, H19 binds to the major histocompatibility complex class II (MHC class II), which facilitates viral entry. Unlike the broad MHC class II specificity of H17 and H18 from bat IAV, H19 exhibits a species-specific MHC class II usage that suggests a limited host range and zoonotic potential. Using cell lines overexpressing MHC class II, we rescued recombinant H19 IAV. We solved the H19 crystal structure and identified residues within the putative Sia receptor binding site (RBS) that impede Sia-dependent entry.
Collapse
Affiliation(s)
- Umut Karakus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA
| | - Jithesh Kottur
- Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sara S El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rocío Seoane
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Soner Yildiz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Leanne Chen
- Department of Biology, Barnard College, New York, NY 10027, USA
| | - Magdalena Plancarte
- Department of Pathology, Microbiology, and Immunology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | - LeAnn Lindsay
- Department of Pathology, Microbiology, and Immunology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | | | | | | | - Geert-Jan Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands; Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA 30602, USA; Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CH Utrecht, the Netherlands; Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Silke Stertz
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Walter Boyce
- Department of Pathology, Microbiology, and Immunology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | - Robert P de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Aneel K Aggarwal
- Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
9
|
Nelli RK, Harm TA, Siepker C, Groeltz-Thrush JM, Jones B, Twu NC, Nenninger AS, Magstadt DR, Burrough ER, Piñeyro PE, Mainenti M, Carnaccini S, Plummer PJ, Bell TM. Sialic Acid Receptor Specificity in Mammary Gland of Dairy Cattle Infected with Highly Pathogenic Avian Influenza A(H5N1) Virus. Emerg Infect Dis 2024; 30:1361-1373. [PMID: 38861554 PMCID: PMC11210646 DOI: 10.3201/eid3007.240689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024] Open
Abstract
In March 2024, the US Department of Agriculture's Animal and Plant Health Inspection Service reported detection of highly pathogenic avian influenza (HPAI) A(H5N1) virus in dairy cattle in the United States for the first time. One factor that determines susceptibility to HPAI H5N1 infection is the presence of specific virus receptors on host cells; however, little is known about the distribution of the sialic acid (SA) receptors in dairy cattle, particularly in mammary glands. We compared the distribution of SA receptors in the respiratory tract and mammary gland of dairy cattle naturally infected with HPAI H5N1. The respiratory and mammary glands of HPAI H5N1-infected dairy cattle are rich in SA, particularly avian influenza virus-specific SA α2,3-gal. Mammary gland tissues co-stained with sialic acids and influenza A virus nucleoprotein showed predominant co-localization with the virus and SA α2,3-gal. HPAI H5N1 exhibited epitheliotropism within the mammary gland, and we observed rare immunolabeling within macrophages.
Collapse
|
10
|
Gadzhiev A, Petherbridge G, Sharshov K, Sobolev I, Alekseev A, Gulyaeva M, Litvinov K, Boltunov I, Teymurov A, Zhigalin A, Daudova M, Shestopalov A. Pinnipeds and avian influenza: a global timeline and review of research on the impact of highly pathogenic avian influenza on pinniped populations with particular reference to the endangered Caspian seal ( Pusa caspica). Front Cell Infect Microbiol 2024; 14:1325977. [PMID: 39071164 PMCID: PMC11273096 DOI: 10.3389/fcimb.2024.1325977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/21/2024] [Indexed: 07/30/2024] Open
Abstract
This study reviews chronologically the international scientific and health management literature and resources relating to impacts of highly pathogenic avian influenza (HPAI) viruses on pinnipeds in order to reinforce strategies for the conservation of the endangered Caspian seal (Pusa caspica), currently under threat from the HPAI H5N1 subtype transmitted from infected avifauna which share its haul-out habitats. Many cases of mass pinniped deaths globally have occurred from HPAI spill-overs, and are attributed to infected sympatric aquatic avifauna. As the seasonal migrations of Caspian seals provide occasions for contact with viruses from infected migratory aquatic birds in many locations around the Caspian Sea, this poses a great challenge to seal conservation. These are thus critical locations for the surveillance of highly pathogenic influenza A viruses, whose future reassortments may present a pandemic threat to humans.
Collapse
Affiliation(s)
- Alimurad Gadzhiev
- Institute of Ecology and Sustainable Development, Dagestan State University, Makhachkala, Russia
| | - Guy Petherbridge
- Institute of Ecology and Sustainable Development, Dagestan State University, Makhachkala, Russia
- Caspian Centre for Nature Conservation, International Institute of Ecology and Sustainable Development, Association of Universities and Research Centers of Caspian Region States, Makhachkala, Russia
| | - Kirill Sharshov
- Research Institute of Virology, Federal Research Centre for Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | - Ivan Sobolev
- Research Institute of Virology, Federal Research Centre for Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Alekseev
- Institute of Ecology and Sustainable Development, Dagestan State University, Makhachkala, Russia
- Research Institute of Virology, Federal Research Centre for Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | - Marina Gulyaeva
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Kirill Litvinov
- Laboratory of Ecological and Biological Research, Astrakhan State Nature Biosphere Reserve, Astrakhan, Russia
| | - Ivan Boltunov
- Department of Vertebrate Zoology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Abdulgamid Teymurov
- Institute of Ecology and Sustainable Development, Dagestan State University, Makhachkala, Russia
| | - Alexander Zhigalin
- Institute of Ecology and Sustainable Development, Dagestan State University, Makhachkala, Russia
| | - Madina Daudova
- Institute of Ecology and Sustainable Development, Dagestan State University, Makhachkala, Russia
| | - Alexander Shestopalov
- Research Institute of Virology, Federal Research Centre for Fundamental and Translational Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
11
|
Tan CCS, van Dorp L, Balloux F. The evolutionary drivers and correlates of viral host jumps. Nat Ecol Evol 2024; 8:960-971. [PMID: 38528191 DOI: 10.1038/s41559-024-02353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 03/27/2024]
Abstract
Most emerging and re-emerging infectious diseases stem from viruses that naturally circulate in non-human vertebrates. When these viruses cross over into humans, they can cause disease outbreaks, epidemics and pandemics. While zoonotic host jumps have been extensively studied from an ecological perspective, little attention has gone into characterizing the evolutionary drivers and correlates underlying these events. To address this gap, we harnessed the entirety of publicly available viral genomic data, employing a comprehensive suite of network and phylogenetic analyses to investigate the evolutionary mechanisms underpinning recent viral host jumps. Surprisingly, we find that humans are as much a source as a sink for viral spillover events, insofar as we infer more viral host jumps from humans to other animals than from animals to humans. Moreover, we demonstrate heightened evolution in viral lineages that involve putative host jumps. We further observe that the extent of adaptation associated with a host jump is lower for viruses with broader host ranges. Finally, we show that the genomic targets of natural selection associated with host jumps vary across different viral families, with either structural or auxiliary genes being the prime targets of selection. Collectively, our results illuminate some of the evolutionary drivers underlying viral host jumps that may contribute to mitigating viral threats across species boundaries.
Collapse
Affiliation(s)
- Cedric C S Tan
- UCL Genetics Institute, University College London, London, UK.
- The Francis Crick Institute, London, UK.
| | - Lucy van Dorp
- UCL Genetics Institute, University College London, London, UK
| | | |
Collapse
|
12
|
Maya-Badillo BA, Orta-Pineda G, Zavala-Vasco D, Rivera-Rosas KE, Uribe-Jacinto A, Segura-Velásquez R, Suzán G, Sánchez-Betancourt JI. Influenza A virus antibodies in dogs, hunting dogs, and backyard pigs in Campeche, Mexico. Zoonoses Public Health 2024; 71:294-303. [PMID: 38196021 DOI: 10.1111/zph.13110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
AIMS This study aimed to identify exposure to human, swine, and avian influenza A virus subtypes in rural companion and hunting dogs, backyard pigs, and feral pigs. METHODS AND RESULTS The study took place in a region of southeastern Mexico where the sampled individuals were part of backyard production systems in which different domestic and wild species coexist and interact with humans. We collected blood samples from pigs and dogs at each of the sites. We used a nucleoprotein enzyme-linked immunosorbent assay to determine the exposure of individuals to influenza A virus. Haemagglutination inhibition was performed on the positive samples to determine the subtypes to which they were exposed. For data analysis, a binomial logistic regression model was generated to determine the predictor variables for the seropositivity of the individuals in the study. We identified 11 positive individuals: three backyard pigs, four companion dogs, and four hunting dogs. The pigs tested positive for H1N1 and H1N2. The dogs were positive for H1N1, H1N2, and H3N2. The model showed that dogs in contact with backyard chickens are more likely to be seropositive for influenza A viruses. CONCLUSIONS We demonstrated the essential role hunting dogs could play as intermediate hosts and potential mixing vessel hosts when exposed to human and swine-origin viral subtypes. These results are relevant because these dogs interact with domestic hosts and humans in backyard systems, which are risk scenarios in the transmission of influenza A viruses. Therefore, it is of utmost importance to implement epidemiological surveillance of influenza A viruses in backyard animals, particularly in key animals in the transmission of these viruses, such as dogs and pigs.
Collapse
Affiliation(s)
- Brenda Aline Maya-Badillo
- Laboratorio de Ecología de Enfermedades y Una Salud, Departamento de Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Grupo de Investigación del Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guillermo Orta-Pineda
- Laboratorio de Ecología de Enfermedades y Una Salud, Departamento de Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diego Zavala-Vasco
- Grupo de Investigación del Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Karen Elizabeth Rivera-Rosas
- Grupo de Investigación del Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - René Segura-Velásquez
- Unidad de Investigación de la Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Suzán
- Laboratorio de Ecología de Enfermedades y Una Salud, Departamento de Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Iván Sánchez-Betancourt
- Grupo de Investigación del Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
13
|
Nilsson J, Eriksson P, Naguib MM, Jax E, Sihlbom C, Olsson BM, Lundkvist Å, Olsen B, Järhult JD, Larson G, Ellström P. Expression of influenza A virus glycan receptor candidates in mallard, chicken, and tufted duck. Glycobiology 2024; 34:cwad098. [PMID: 38127648 PMCID: PMC10987293 DOI: 10.1093/glycob/cwad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Influenza A virus (IAV) pandemics result from interspecies transmission events within the avian reservoir and further into mammals including humans. Receptor incompatibility due to differently expressed glycan structures between species has been suggested to limit zoonotic IAV transmission from the wild bird reservoir as well as between different bird species. Using glycoproteomics, we have studied the repertoires of expressed glycan structures with focus on putative sialic acid-containing glycan receptors for IAV in mallard, chicken and tufted duck; three bird species with different roles in the zoonotic ecology of IAV. The methodology used pinpoints specific glycan structures to specific glycosylation sites of identified glycoproteins and was also used to successfully discriminate α2-3- from α2-6-linked terminal sialic acids by careful analysis of oxonium ions released from glycopeptides in tandem MS/MS (MS2), and MS/MS/MS (MS3). Our analysis clearly demonstrated that all three bird species can produce complex N-glycans including α2-3-linked sialyl Lewis structures, as well as both N- and O- glycans terminated with both α2-3- and α2-6-linked Neu5Ac. We also found the recently identified putative IAV receptor structures, Man-6P N-glycopeptides, in all tissues of the three bird species. Furthermore, we found many similarities in the repertoires of expressed receptors both between the bird species investigated and to previously published data from pigs and humans. Our findings of sialylated glycan structures, previously anticipated to be mammalian specific, in all three bird species may have major implications for our understanding of the role of receptor incompatibility in interspecies transmission of IAV.
Collapse
Affiliation(s)
- Jonas Nilsson
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Vita Stråket 12, Gothenburg SE-413 45, Sweden
- Laboratory of Clinical Chemistry, Sahlgrenska University Hospital, Bruna Stråket 16, Gothenburg SE-413 45, Sweden
- Proteomics Core Facility, University of Gothenburg, Sahlgrenska Academy, Medicinaregatan 9E, Gothenburg SE-405 30, Sweden
| | - Per Eriksson
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| | - Mahmoud M Naguib
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Husargatan 3, Uppsala University, Uppsala, SE-75237, Sweden
| | - Elinor Jax
- Department of Migration, Max Planck Institute of Animal Behavior, Am Obstberg 1, Radolfzell, Baden-Württemberg DE-78315, Germany
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Sahlgrenska Academy, Medicinaregatan 9E, Gothenburg SE-405 30, Sweden
| | - Britt-Marie Olsson
- Proteomics Core Facility, University of Gothenburg, Sahlgrenska Academy, Medicinaregatan 9E, Gothenburg SE-405 30, Sweden
| | - Åke Lundkvist
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Husargatan 3, Uppsala University, Uppsala, SE-75237, Sweden
| | - Björn Olsen
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| | - Josef D Järhult
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Vita Stråket 12, Gothenburg SE-413 45, Sweden
- Laboratory of Clinical Chemistry, Sahlgrenska University Hospital, Bruna Stråket 16, Gothenburg SE-413 45, Sweden
| | - Patrik Ellström
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| |
Collapse
|
14
|
Pavia G, Scarpa F, Ciccozzi A, Romano C, Branda F, Quirino A, Marascio N, Matera G, Sanna D, Ciccozzi M. Changing and Evolution of Influenza Virus: Is It a Trivial Flu? Chemotherapy 2024; 69:185-193. [PMID: 38508151 DOI: 10.1159/000538382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Influenza viruses are etiological agents which cause contagious respiratory, seasonal epidemics and, for influenza A subtypes, pandemics. The clinical picture of influenza has undergone continuous change over the years, due to intrinsic viral evolution as well as "reassortment" of its genomic segments. The history of influenza highlights its ability to adapt and to rapidly evolve, without specific circumstances. This reflects the complexity of this pathology and poses the fundamental question about its assumption as a "common illness" and its impact on public health. SUMMARY The global influenza epidemics and pandemics claimed millions of deaths, leaving an indelible mark on public health and showing the need for a better comprehension of the influenza virus. The clear understanding of genetic variations during the influenza seasonal epidemics is a crucial point for developing effective strategies for prevention, treatment, and vaccine design. The recent advance in next-generation sequencing approaches, model systems to virus culture, and bioinformatics pipeline played a key role in the rapid characterization of circulating influenza strains. In particular, the increase in computational power allowed the performance of complex tasks in healthcare settings through machine learning algorithms, which analyze different variables, such as medical and laboratory outputs, to optimize medical research and improve public health systems. The early detection of emerging and reemerging pathogens is a matter of importance to prevent future pandemics. KEY MESSAGES The perception of influenza as a "trivial flu" or a more serious public health concern is a subject of ongoing debate, reflecting the multifaceted nature of this infectious disease. The variability in the severity of influenza sheds light on the unpredictability of the viral characteristics, coupled with the challenges in accurately predicting circulating strains. This adds complexity to the public health burden of influenza and highlights the need for targeted interventions.
Collapse
Affiliation(s)
- Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
15
|
Kumar G, Sakharam KA. Tackling Influenza A virus by M2 ion channel blockers: Latest progress and limitations. Eur J Med Chem 2024; 267:116172. [PMID: 38330869 DOI: 10.1016/j.ejmech.2024.116172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024]
Abstract
Influenza outbreaks cause pandemics in millions of people. The treatment of influenza remains a challenge due to significant genetic polymorphism in the influenza virus. Also, developing vaccines to protect against seasonal and pandemic influenza infections is constantly impeded. Thus, antibiotics are the only first line of defense against antigenically distinct strains or new subtypes of influenza viruses. Among several anti-influenza targets, the M2 protein of the influenza virus performs several activities. M2 protein is an ion channel that permits proton conductance through the virion envelope and the deacidification of the Golgi apparatus. Both these functions are critical for viral replication. Thus, targeting the M2 protein of the influenza virus is an essential target. Rimantadine and amantadine are two well-known drugs that act on the M2 protein. However, these drugs acquired resistance to influenza and thus are not recommended to treat influenza infections. This review discusses an overview of anti-influenza therapy, M2 ion channel functions, and its working principle. It also discusses the M2 structure and its role, and the change in the structure leads to mutant variants of influenza A virus. We also shed light on the recently identified compounds acting against wild-type and mutated M2 proteins of influenza virus A. These scaffolds could be an alternative to M2 inhibitors and be developed as antibiotics for treating influenza infections.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India.
| | - Kakade Aditi Sakharam
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| |
Collapse
|
16
|
Wang Q, Liu Z, Zeng X, Zheng Y, Lan L, Wang X, Lai Z, Hou X, Gao L, Liang L, Tang S, Zhang Z, Leng J, Fan X. Integrated analysis of miRNA-mRNA expression of newly emerging swine H3N2 influenza virus cross-species infection with tree shrews. Virol J 2024; 21:4. [PMID: 38178220 PMCID: PMC10768296 DOI: 10.1186/s12985-023-02260-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Cross-species transmission of zoonotic IAVs to humans is potentially widespread and lethal, posing a great threat to human health, and their cross-species transmission mechanism has attracted much attention. miRNAs have been shown to be involved in the regulation of IAVs infection and immunity, however, few studies have focused on the molecular mechanisms underlying miRNAs and mRNAs expression after IAVs cross-species infection. METHODS We used tree shrews, a close relative of primates, as a model and used RNA-Seq and bioinformatics tools to analyze the expression profiles of DEMs and DEGs in the nasal turbinate tissue at different time points after the newly emerged swine influenza A virus SW2783 cross-species infection with tree shrews, and miRNA-mRNA interaction maps were constructed and verified by RT-qPCR, miRNA transfection and luciferase reporter assay. RESULTS 14 DEMs were screened based on functional analysis and interaction map, miR-760-3p, miR-449b-2, miR-30e-3p, and miR-429 were involved in the signal transduction process of replication and proliferation after infection, miR-324-3p, miR-1301-1, miR-103-1, miR-134-5p, miR-29a, miR-31, miR-16b, miR-34a, and miR-125b participate in negative feedback regulation of genes related to the immune function of the body to activate the antiviral immune response, and miR-106b-3p may be related to the cross-species infection potential of SW2783, and the expression level of these miRNAs varies in different days after infection. CONCLUSIONS The miRNA regulatory networks were constructed and 14 DEMs were identified, some of them can affect the replication and proliferation of viruses by regulating signal transduction, while others can play an antiviral role by regulating the immune response. It indicates that abnormal expression of miRNAs plays a crucial role in the regulation of cross-species IAVs infection, which lays a solid foundation for further exploration of the molecular regulatory mechanism of miRNAs in IAVs cross-species infection and anti-influenza virus targets.
Collapse
Affiliation(s)
- Qihui Wang
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China.
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| | - Zihe Liu
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
| | - Xia Zeng
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yu Zheng
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
| | - Li Lan
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
| | - Xinhang Wang
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
| | - Zhenping Lai
- Department of Microbiology, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoqiong Hou
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
| | - Lingxi Gao
- Department of Microbiology, Guangxi Medical University, Nanning, 530021, China
| | - Liang Liang
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Shen Tang
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Zengfeng Zhang
- Department of Microbiology, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Jing Leng
- Department of Immunology, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China.
- Key Laboratory of Characteristic Experimental Animal Models of Guangxi, Guangxi University of Chinese Medicine, Nanning, 530200, China.
| | - Xiaohui Fan
- Department of Microbiology, Guangxi Medical University, Nanning, 530021, China.
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| |
Collapse
|
17
|
Bianconi I, Aschbacher R, Pagani E. Current Uses and Future Perspectives of Genomic Technologies in Clinical Microbiology. Antibiotics (Basel) 2023; 12:1580. [PMID: 37998782 PMCID: PMC10668849 DOI: 10.3390/antibiotics12111580] [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: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
Recent advancements in sequencing technology and data analytics have led to a transformative era in pathogen detection and typing. These developments not only expedite the process, but also render it more cost-effective. Genomic analyses of infectious diseases are swiftly becoming the standard for pathogen analysis and control. Additionally, national surveillance systems can derive substantial benefits from genomic data, as they offer profound insights into pathogen epidemiology and the emergence of antimicrobial-resistant strains. Antimicrobial resistance (AMR) is a pressing global public health issue. While clinical laboratories have traditionally relied on culture-based antimicrobial susceptibility testing, the integration of genomic data into AMR analysis holds immense promise. Genomic-based AMR data can furnish swift, consistent, and highly accurate predictions of resistance phenotypes for specific strains or populations, all while contributing invaluable insights for surveillance. Moreover, genome sequencing assumes a pivotal role in the investigation of hospital outbreaks. It aids in the identification of infection sources, unveils genetic connections among isolates, and informs strategies for infection control. The One Health initiative, with its focus on the intricate interconnectedness of humans, animals, and the environment, seeks to develop comprehensive approaches for disease surveillance, control, and prevention. When integrated with epidemiological data from surveillance systems, genomic data can forecast the expansion of bacterial populations and species transmissions. Consequently, this provides profound insights into the evolution and genetic relationships of AMR in pathogens, hosts, and the environment.
Collapse
Affiliation(s)
- Irene Bianconi
- Laboratory of Microbiology and Virology, Provincial Hospital of Bolzano (SABES-ASDAA), Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversitätvia Amba Alagi 5, 39100 Bolzano, Italy; (R.A.); (E.P.)
| | | | | |
Collapse
|
18
|
Godoy M, de Oca MM, Caro D, Pontigo JP, Kibenge M, Kibenge F. Evolution and Current Status of Influenza A Virus in Chile: A Review. Pathogens 2023; 12:1252. [PMID: 37887768 PMCID: PMC10610240 DOI: 10.3390/pathogens12101252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
The influenza A virus (IAV) poses a significant global threat to public health and food security. Particularly concerning is the avian influenza virus (AIV) subtype H5N1, which has spread from Europe to North and Central/South America. This review presents recent developments in IAV evolution in birds, mammals, and humans in Chile. Chile's encounter with IAV began in 2002, with the highly pathogenic avian influenza (HPAI) H7N3 virus, derived from a unique South American low pathogenic avian influenza (LPAI) virus. In 2016-2017, LPAI H7N6 caused outbreaks in turkey, linked to wild birds in Chile and Bolivia. The pandemic influenza A (H1N1) 2009 (H1N1pdm09) virus in 2009 decreased egg production in turkeys. Since 2012, diverse IAV subtypes have emerged in backyard poultry and pigs. Reassortant AIVs, incorporating genes from both North and South American isolates, have been found in wild birds since 2007. Notably, from December 2022, HPAI H5N1 was detected in wild birds, sea lions, and a human, along Chile's north coast. It was introduced through Atlantic migratory flyways from North America. These findings emphasize the need for enhanced biosecurity on poultry farms and ongoing genomic surveillance to understand and manage AIVs in both wild and domestic bird populations in Chile.
Collapse
Affiliation(s)
- Marcos Godoy
- Centro de Investigaciones Biológicas Aplicadas (CIBA), Puerto Montt 5480000, Chile; (M.M.d.O.); (D.C.)
- Laboratorio de Biotecnología Aplicada, Facultad de Ciencias de la Naturaleza, Escuela de Medicina Veterinaria, Sede de la Patagonia, Universidad San Sebastián, Puerto Montt 5480000, Chile;
| | - Marco Montes de Oca
- Centro de Investigaciones Biológicas Aplicadas (CIBA), Puerto Montt 5480000, Chile; (M.M.d.O.); (D.C.)
| | - Diego Caro
- Centro de Investigaciones Biológicas Aplicadas (CIBA), Puerto Montt 5480000, Chile; (M.M.d.O.); (D.C.)
| | - Juan Pablo Pontigo
- Laboratorio de Biotecnología Aplicada, Facultad de Ciencias de la Naturaleza, Escuela de Medicina Veterinaria, Sede de la Patagonia, Universidad San Sebastián, Puerto Montt 5480000, Chile;
| | - Molly Kibenge
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, 550 University Ave, Charlottetown, PE C1A 4P3, Canada;
| | - Frederick Kibenge
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, 550 University Ave, Charlottetown, PE C1A 4P3, Canada;
| |
Collapse
|
19
|
Chow VTK, Tay DJW, Chen MIC, Tang JW, Milton DK, Tham KW. Influenza A and B Viruses in Fine Aerosols of Exhaled Breath Samples from Patients in Tropical Singapore. Viruses 2023; 15:2033. [PMID: 37896810 PMCID: PMC10612062 DOI: 10.3390/v15102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Influenza is a highly contagious respiratory illness that commonly causes outbreaks among human communities. Details about the exact nature of the droplets produced by human respiratory activities such as breathing, and their potential to carry and transmit influenza A and B viruses is still not fully understood. The objective of our study was to characterize and quantify influenza viral shedding in exhaled aerosols from natural patient breath, and to determine their viral infectivity among participants in a university cohort in tropical Singapore. Using the Gesundheit-II exhaled breath sampling apparatus, samples of exhaled breath of two aerosol size fractions ("coarse" > 5 µm and "fine" ≤ 5 µm) were collected and analyzed from 31 study participants, i.e., 24 with influenza A (including H1N1 and H3N2 subtypes) and 7 with influenza B (including Victoria and Yamagata lineages). Influenza viral copy number was quantified using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Infectivity of influenza virus in the fine particle fraction was determined by culturing in Madin-Darby canine kidney cells. Exhaled influenza virus RNA generation rates ranged from 9 to 1.67 × 105 and 10 to 1.24 × 104 influenza virus RNA copies per minute for the fine and coarse aerosol fractions, respectively. Compared to the coarse aerosol fractions, influenza A and B viruses were detected more frequently in the fine aerosol fractions that harbored 12-fold higher viral loads. Culturable virus was recovered from the fine aerosol fractions from 9 of the 31 subjects (29%). These findings constitute additional evidence to reiterate the important role of fine aerosols in influenza transmission and provide a baseline range of influenza virus RNA generation rates.
Collapse
Affiliation(s)
- Vincent T. K. Chow
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
| | - Douglas Jie Wen Tay
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
| | - Mark I. C. Chen
- Research Office, National Centre for Infectious Diseases, Singapore 308442, Singapore;
| | - Julian W. Tang
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 7RH, UK;
| | - Donald K. Milton
- Institute for Applied Environmental Health, University of Maryland School of Public Health, College Park, MD 20742, USA;
| | - Kwok Wai Tham
- Department of the Built Environment, College of Design and Engineering, National University of Singapore, Singapore 117356, Singapore
| |
Collapse
|
20
|
van Diemen PM, Byrne AMP, Ramsay AM, Watson S, Nunez A, V Moreno A, Chiapponi C, Foni E, Brown IH, Brookes SM, Everett HE. Interspecies Transmission of Swine Influenza A Viruses and Human Seasonal Vaccine-Mediated Protection Investigated in Ferret Model. Emerg Infect Dis 2023; 29:1798-1807. [PMID: 37610158 PMCID: PMC10461666 DOI: 10.3201/eid2909.230066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
We investigated the infection dynamics of 2 influenza A(H1N1) virus isolates from the swine 1A.3.3.2 (pandemic 2009) and 1C (Eurasian, avian-like) lineages. The 1C-lineage virus, A/Pavia/65/2016, although phylogenetically related to swine-origin viruses, was isolated from a human clinical case. This strain infected ferrets, a human influenza model species, and could be transmitted by direct contact and, less efficiently, by airborne exposure. Infecting ferrets and pigs (the natural host) resulted in mild or inapparent clinical signs comparable to those observed with 1A.3.3.2-lineage swine-origin viruses. Both H1N1 viruses could infect pigs and were transmitted to cohoused ferrets. Ferrets vaccinated with a human 2016-17 seasonal influenza vaccine were protected against infection with the antigenically matched 1A pandemic 2009 virus but not against the swine-lineage 1C virus. Our results reaffirm the need for continuous influenza A virus surveillance in pigs and identification of candidate human vaccine viruses.
Collapse
|
21
|
Naguib MM, Eriksson P, Jax E, Wille M, Lindskog C, Bröjer C, Krambrich J, Waldenström J, Kraus RHS, Larson G, Lundkvist Å, Olsen B, Järhult JD, Ellström P. A Comparison of Host Responses to Infection with Wild-Type Avian Influenza Viruses in Chickens and Tufted Ducks. Microbiol Spectr 2023; 11:e0258622. [PMID: 37358408 PMCID: PMC10434033 DOI: 10.1128/spectrum.02586-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 05/31/2023] [Indexed: 06/27/2023] Open
Abstract
Cross-species transmission of influenza A virus (IAV) from wild waterfowl to poultry is the first step in a chain of events that can ultimately lead to exposure and infection of humans. Herein, we study the outcome of infection with eight different mallard-origin IAV subtypes in two different avian hosts: tufted ducks and chickens. We found that infection and shedding patterns as well as innate immune responses were highly dependent on viral subtypes, host species, and inoculation routes. For example, intraoesophageal inoculation, commonly used in mallard infection experiments, resulted in no infections in contrast to oculonasal inoculation, suggesting a difference in transmission routes. Despite H9N2 being endemic in chickens, inoculation of mallard-origin H9N2 failed to cause viable infection beyond 1 day postinfection in our study design. The innate immune responses were markedly different in chickens and tufted ducks, and despite the presence of retinoic acid-inducible gene-I (RIG-I) in tufted duck transcriptomes, it was neither up nor downregulated in response to infection. Overall, we have revealed the heterogeneity of infection patterns and responses in two markedly different avian hosts following a challenge with mallard-origin IAV. These virus-host interactions provide new insights into important aspects of interspecies transmission of IAV. IMPORTANCE Our current findings highlight important aspects of IAV infection in birds that have implications for our understanding of its zoonotic ecology. In contrast to mallards where the intestinal tract is the main site of IAV replication, chickens and tufted ducks show limited or no signs of intestinal infection suggesting that the fecal-oral transmission route might not apply to all bird IAV host species. Our results indicate that mallard-origin IAVs undergo genetic changes upon introduction into new hosts, suggesting rapid adaptation to a new environment. However, similar to the mallard, chickens and tufted ducks show a limited immune response to infection with low pathogenic avian influenza viruses. These findings and future studies in different IAV hosts are important for our understanding of barriers to IAV transmission between species and ultimately from the wild reservoir to humans.
Collapse
Affiliation(s)
- Mahmoud M. Naguib
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Per Eriksson
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Elinor Jax
- Department of Migration, Max Planck Institute of Animal Behavior, Radolfzell, Germany
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Michelle Wille
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Caroline Bröjer
- Department of Pathology and Wildlife Diseases, National Veterinary Institute (SVA), Uppsala, Sweden
| | - Janina Krambrich
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jonas Waldenström
- Centre for Ecology and Evolution in Microbial Model Systems, Linnaeus University, Kalmar, Sweden
| | - Robert H. S. Kraus
- Department of Migration, Max Planck Institute of Animal Behavior, Radolfzell, Germany
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Göran Larson
- Department of Laboratory Medicine, University of Gothenburg, Gothenburg, Sweden
- Laboratory of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Åke Lundkvist
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Björn Olsen
- Zoonosis Science Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Josef D. Järhult
- Zoonosis Science Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Patrik Ellström
- Zoonosis Science Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Pinto RM, Bakshi S, Lytras S, Zakaria MK, Swingler S, Worrell JC, Herder V, Hargrave KE, Varjak M, Cameron-Ruiz N, Collados Rodriguez M, Varela M, Wickenhagen A, Loney C, Pei Y, Hughes J, Valette E, Turnbull ML, Furnon W, Gu Q, Orr L, Taggart A, Diebold O, Davis C, Boutell C, Grey F, Hutchinson E, Digard P, Monne I, Wootton SK, MacLeod MKL, Wilson SJ, Palmarini M. BTN3A3 evasion promotes the zoonotic potential of influenza A viruses. Nature 2023; 619:338-347. [PMID: 37380775 DOI: 10.1038/s41586-023-06261-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.
Collapse
Affiliation(s)
- Rute Maria Pinto
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Siddharth Bakshi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Spyros Lytras
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Simon Swingler
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Julie C Worrell
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Kerrie E Hargrave
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Margus Varjak
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- Faculty of Science and Technology, Institute of Technology, University of Tartu, Tartu, Estonia
| | | | | | - Mariana Varela
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Colin Loney
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Elise Valette
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Wilhelm Furnon
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Lauren Orr
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Aislynn Taggart
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Ola Diebold
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Chris Davis
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Chris Boutell
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Finn Grey
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | | | - Paul Digard
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Isabella Monne
- Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), Legnaro, Italy
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Megan K L MacLeod
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Sam J Wilson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | |
Collapse
|
23
|
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: 1.0] [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
|
24
|
Wasberg A, Faria IR, Bergholm J, Petric PP, Mostafa A, Pleschka S, Schwemmle M, Lundkvist Å, Ellström P, Naguib MM. Assessing compatibility and viral fitness between poultry-adapted H9N2 and wild bird-derived neuraminidases. Sci Rep 2023; 13:4476. [PMID: 36934147 PMCID: PMC10024770 DOI: 10.1038/s41598-023-31653-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Exchange of viral segments between one or more influenza virus subtypes can contribute to a shift in virulence and adaptation to new hosts. Among several influenza subtypes, H9N2 is widely circulating in poultry populations worldwide and has the ability to infect humans. Here, we studied the reassortant compatibility between chicken H9N2 with N1-N9 gene segments of wild bird origin, either with an intact or truncated stalk. Naturally occurring amino acid deletions in the NA stalk of the influenza virus can lead to increased virulence in both mallard ducks and chickens. Our findings show extended genetic compatibility between chicken H9Nx gene segments and the wild-bird NA with and without 20 amino acid stalk deletion. Replication kinetics in avian, mammalian and human cell lines revealed that parental chH9N2 and rH9N6 viruses with intact NA-stalk replicated significantly better in avian DF1 cells compared to human A549 cells. After introducing a stalk deletion, an enhanced preference for replication in mammalian and human cell lines could be observed for rH9N2Δ(H6), rH9N6Δ and rH9N9Δ compared to the parental chH9N2 virus. This highlights the potential emergence of novel viruses with variable phenotypic traits, warranting the continuous monitoring of H9N2 and co-circulating subtypes in avian hosts.
Collapse
Affiliation(s)
- Anishia Wasberg
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Inês R Faria
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Julia Bergholm
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, 750 07, Uppsala, Sweden
| | - Philipp P Petric
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Stephan Pleschka
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany
- German Center for Infection Research (DZIF),partner site Giessen-Marburg-Langen, Giessen, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Åke Lundkvist
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Patrik Ellström
- Zoonosis Science Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mahmoud M Naguib
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
25
|
Souza CK, Kimble JB, Anderson TK, Arendsee ZW, Hufnagel DE, Young KM, Gauger PC, Lewis NS, Davis CT, Thor S, Vincent Baker AL. Swine-to-Ferret Transmission of Antigenically Drifted Contemporary Swine H3N2 Influenza A Virus Is an Indicator of Zoonotic Risk to Humans. Viruses 2023; 15:v15020331. [PMID: 36851547 PMCID: PMC9962742 DOI: 10.3390/v15020331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023] Open
Abstract
Human-to-swine transmission of influenza A (H3N2) virus occurs repeatedly and plays a critical role in swine influenza A virus (IAV) evolution and diversity. Human seasonal H3 IAVs were introduced from human-to-swine in the 1990s in the United States and classified as 1990.1 and 1990.4 lineages; the 1990.4 lineage diversified into 1990.4.A-F clades. Additional introductions occurred in the 2010s, establishing the 2010.1 and 2010.2 lineages. Human zoonotic cases with swine IAV, known as variant viruses, have occurred from the 1990.4 and 2010.1 lineages, highlighting a public health concern. If a variant virus is antigenically drifted from current human seasonal vaccine (HuVac) strains, it may be chosen as a candidate virus vaccine (CVV) for pandemic preparedness purposes. We assessed the zoonotic risk of US swine H3N2 strains by performing phylogenetic analyses of recent swine H3 strains to identify the major contemporary circulating genetic clades. Representatives were tested in hemagglutination inhibition assays with ferret post-infection antisera raised against existing CVVs or HuVac viruses. The 1990.1, 1990.4.A, and 1990.4.B.2 clade viruses displayed significant loss in cross-reactivity to CVV and HuVac antisera, and interspecies transmission potential was subsequently investigated in a pig-to-ferret transmission study. Strains from the three lineages were transmitted from pigs to ferrets via respiratory droplets, but there were differential shedding profiles. These data suggest that existing CVVs may offer limited protection against swine H3N2 infection, and that contemporary 1990.4.A viruses represent a specific concern given their widespread circulation among swine in the United States and association with multiple zoonotic cases.
Collapse
Affiliation(s)
- Carine K. Souza
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
| | - J. Brian Kimble
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
| | - Tavis K. Anderson
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
| | - Zebulun W. Arendsee
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
| | - David E. Hufnagel
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
| | - Katharine M. Young
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
| | - Phillip C. Gauger
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Nicola S. Lewis
- Department of Pathology and Population Sciences, Royal Veterinary College, University of London, Hertfordshire, London NW1 0TU, UK
| | - C. Todd Davis
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Sharmi Thor
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Amy L. Vincent Baker
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Ames, IA 50010, USA
- Correspondence:
| |
Collapse
|
26
|
Braun KM, Haddock III LA, Crooks CM, Barry GL, Lalli J, Neumann G, Watanabe T, Imai M, Yamayoshi S, Ito M, Moncla LH, Koelle K, Kawaoka Y, Friedrich TC. Avian H7N9 influenza viruses are evolutionarily constrained by stochastic processes during replication and transmission in mammals. Virus Evol 2023; 9:vead004. [PMID: 36814938 PMCID: PMC9939568 DOI: 10.1093/ve/vead004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
H7N9 avian influenza viruses (AIVs) have caused over 1,500 documented human infections since emerging in 2013. Although wild-type H7N9 AIVs can be transmitted by respiratory droplets in ferrets, they have not yet caused widespread outbreaks in humans. Previous studies have revealed molecular determinants of H7N9 AIV host switching, but little is known about potential evolutionary constraints on this process. Here, we compare patterns of sequence evolution for H7N9 AIV and mammalian H1N1 viruses during replication and transmission in ferrets. We show that three main factors-purifying selection, stochasticity, and very narrow transmission bottlenecks-combine to severely constrain the ability of H7N9 AIV to effectively adapt to mammalian hosts in isolated, acute spillover events. We find rare evidence of natural selection favoring new, potentially mammal-adapting mutations within ferrets but no evidence of natural selection acting during transmission. We conclude that human-adapted H7N9 viruses are unlikely to emerge during typical spillover infections. Our findings are instead consistent with a model in which the emergence of a human-transmissible virus would be a rare and unpredictable, though highly consequential, 'jackpot' event. Strategies to control the total number of spillover infections will limit opportunities for the virus to win this evolutionary lottery.
Collapse
Affiliation(s)
| | | | - Chelsea M Crooks
- AIDS Vaccine Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, 585 Science Dr. Madison, WI 53711, USA
| | - Gabrielle L Barry
- AIDS Vaccine Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, 585 Science Dr. Madison, WI 53711, USA
| | - Joseph Lalli
- Department of Genetics, University of Wisconsin-Madison, 425 Henry Mall Madison, WI 53706, US
| | - Gabriele Neumann
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, 575 Science Dr. Madison, WI 53711, USA
| | - Tokiko Watanabe
- Division of Virology, Institute of Medical Science, University of Tokyo, 4 Chome-6-1 Shirokanedai Minato City, Tokyo 108-0071, Japan,Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka Suita City, Osaka 565-0871, Japan,Center for Infectious Disease Education and Research (CiDER), Osaka University, 2-8 Yamadaoka Suita City, Osaka 565-0871, Japan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, 4 Chome-6-1 Shirokanedai Minato City, Tokyo 108-0071, Japan,The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, 1 Chome-21-1 Toyama Shinjuku City, Tokyo 162-8655, Japan
| | | | - Mutsumi Ito
- Division of Virology, Institute of Medical Science, University of Tokyo, 4 Chome-6-1 Shirokanedai Minato City, Tokyo 108-0071, Japan
| | | | | | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, 575 Science Dr. Madison, WI 53711, USA,Division of Virology, Institute of Medical Science, University of Tokyo, 4 Chome-6-1 Shirokanedai Minato City, Tokyo 108-0071, Japan,The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, 1 Chome-21-1 Toyama Shinjuku City, Tokyo 162-8655, Japan
| | | |
Collapse
|
27
|
Pegg CL, Schulz BL, Neely BA, Albery GF, Carlson CJ. Glycosylation and the global virome. Mol Ecol 2023; 32:37-44. [PMID: 36217579 PMCID: PMC10947461 DOI: 10.1111/mec.16731] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 12/29/2022]
Abstract
The sugars that coat the outsides of viruses and host cells are key to successful disease transmission, but they remain understudied compared to other molecular features. Understanding the comparative zoology of glycosylation - and harnessing it for predictive science - could help close the molecular gap in zoonotic risk assessment.
Collapse
Affiliation(s)
- Cassandra L. Pegg
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Benjamin L. Schulz
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Benjamin A. Neely
- National Institute of Standards and TechnologyCharlestonSouth CarolinaUSA
| | - Gregory F. Albery
- Department of BiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Colin J. Carlson
- Department of BiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
- Department of Microbiology and ImmunologyGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
- Center for Global Health Science and SecurityGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
28
|
Antiviral activity of CAVAC-1901, a combination of 3 standardized medicinal plants, against highly pathogenic influenza A virus in chickens. Poult Sci 2022; 102:102315. [PMID: 36473384 PMCID: PMC9720341 DOI: 10.1016/j.psj.2022.102315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Three different medicinal plants that consisted of the formulated mixture (CAVAC-1901) have been traditionally used for distinct medicinal purposes in different areas. Angelica dahurica has been used as an important ingredient of a prescription, Gumiganghwal-tang, for the common cold and influenza. Curcuma longa has been utilized for the treatment of asthma, and jaundice. Pinus densiflora (Korean red pine) has been used to improve memory and brain function for the treatment of vascular. Industrial livestock, which are characterized by dense breeding, are vulnerable to influenza infection, causing severe economic loss and social problems. However, there are no viable alternatives due to the risk of the occurrence of variants. Therefore, the aim of this study was to discover anti-influenza combinations of different medicinal plants with the concept of a multicomponent and multitarget (MCMT) strategy in traditional Chinese medicine (TCM). As part of a continuous project, 3 medicinal plants whose inhibitory activity against influenza A was previously reported at the compound level, and the inhibition of cytopathic effects (CPEs) by these formulated mixtures was evaluated against influenza A virus H1N1. A selected combination with an optimal ratio exhibiting synergistic activity was assessed for its antiviral activity in chickens against the highly pathogenic avian influenza (HPAI) H5N6. The selected combination (CAVAC-1901) showed potent inhibitory effects on the expression of neuraminidase and nucleoprotein, by RT-qPCR, Western blot, and immunofluorescence assays. The antiviral activity was more evident in chickens infected with H5N6. The sample-treated group (50 mg/kg/d) decreased mortality and virus titers in various organs. Our results indirectly suggest synergistic inhibitory activity of the combination of 3 different medicinal plants with different modes of action. Taken together, an optimally formulated mixture (CAVAC-1901) could serve as an effective alternative to current measures to minimize damage caused by HPAIs.
Collapse
|
29
|
Qian J, Wu Z, Zhu Y, Liu C. One Health: a holistic approach for food safety in livestock. SCIENCE IN ONE HEALTH 2022; 1:100015. [PMID: 39076604 PMCID: PMC11262287 DOI: 10.1016/j.soh.2023.100015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/07/2023] [Indexed: 07/31/2024]
Abstract
The food safety of livestock is a critical issue between animals and humans due to their complex interactions. Pathogens have the potential to spread at every stage of the animal food handling process, including breeding, processing, packaging, storage, transportation, marketing and consumption. In addition, application of the antibiotic usage in domestic animals is a controversial issue because, while they can combat food-borne zoonotic pathogens and promote animal growth and productivity, they can also lead to the transmission of antibiotic-resistant microorganisms and antibiotic-resistant genes across species and habitats. Coevolution of microbiomes may occur in humans and animals as well which may alter the structure of the human microbiome through animal food consumption. One Health is a holistic approach to systematically understand the complex relationships among humans, animals and environments which may provide effective countermeasures to solve food safety problems aforementioned. This paper depicts the main pathogen spectrum of livestock and animal products, summarizes the flow of antibiotic-resistant bacteria and genes between humans and livestock along the food-chain production, and the correlation of their microbiome is reviewed as well to advocate for deeper interdisciplinary communication and collaboration among researchers in medicine, epidemiology, veterinary medicine and ecology to promote One Health approaches to address the global food safety challenges.
Collapse
Affiliation(s)
- Jing Qian
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheyuan Wu
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yongzhang Zhu
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chang Liu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
30
|
Transmission and pathogenicity of canine H3N2 influenza virus in dog and guinea pig models. Virol J 2022; 19:162. [PMID: 36224594 PMCID: PMC9559841 DOI: 10.1186/s12985-022-01888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/17/2022] [Accepted: 09/26/2022] [Indexed: 12/05/2022] Open
Abstract
Background Influenza A virus causes respiratory disease in many animal species as well as in humans. Due to the high human-animal interface, the monitoring of canine influenza in dogs and the study of the transmission and pathogenicity of canine influenza in animals are important. Methods Eight-week-old beagle dogs (Canis lupus familaris) (n = 13) were used for the intraspecies transmission model. The dogs were inoculated intranasally with 1 ml of 106 EID50 per ml of canine H3N2 influenza virus (A/canine/Thailand/CU-DC5299/2012) (CIV-H3N2). In addition, 4-week-old guinea pigs (Cavia porcellus) (n = 20) were used for the interspecies transmission model. The guinea pigs were inoculated intranasally with 300 µl of 106 EID50 per ml of CIV-H3N2. Results For the Thai CIV-H3N2 challenged in the dog model, the incoculated and direct contact dogs developed respiratory signs at 2 dpi. The dogs shed the virus in the respiratory tract at 1 dpi and developed an H3-specific antibody against the virus at 10 dpi. Lung congestion and histopathological changes in the lung were observed. For the Thai CIV-H3N2 challenge in the guinea pig model, the incoculated, direct contact and aerosol-exposed guinea pigs developed fever at 1–2 dpi. The guinea pigs shed virus in the respiratory tract at 2 dpi and developed an H3-specific antibody against the virus at 7 dpi. Mild histopathological changes in the lung were observed. Conclusion The result of this study demonstrated evidence of intraspecies and interspecies transmission of CIV-H3N2 in a mammalian model.
Collapse
|
31
|
Graaf A, Petric PP, Sehl-Ewert J, Henritzi D, Breithaupt A, King J, Pohlmann A, Deutskens F, Beer M, Schwemmle M, Harder T. Cold-passaged isolates and bat-swine influenza a chimeric viruses as modified live-attenuated vaccines against influenza a viruses in pigs. Vaccine 2022; 40:6255-6270. [PMID: 36137904 DOI: 10.1016/j.vaccine.2022.09.013] [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: 07/14/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Swine influenza A virus (swIAV) infections in pig populations cause considerable morbidity and economic losses. Frequent reverse zoonotic incursions of human IAV boost reassortment opportunities with authentic porcine and avian-like IAV in swine herds potentially enhancing zoonotic and even pre-pandemic potential. Vaccination using adjuvanted inactivated full virus vaccines is frequently used in attempting control of swIAV infections. Accelerated antigenic drift of swIAV in large swine holdings and interference of maternal antibodies with vaccine in piglets can compromise these efforts. Potentially more efficacious modified live-attenuated vaccines (MLVs) bear the risk of reversion of MLV to virulence. Here we evaluated new MLV candidates based on cold-passaged swIAV or on reassortment-incompetent bat-IAV-swIAV chimeric viruses. Serial cold-passaging of various swIAV subtypes did not yield unambiguously temperature-sensitive mutants although safety studies in mice and pigs suggested some degree of attenuation. Chimeric bat-swIAV expressing the hemagglutinin and neuraminidase of an avian-like H1N1, in contrast, proved to be safe in mice and pigs, and a single nasal inoculation induced protective immunity against homologous challenge in pigs. Reassortant-incompetent chimeric bat-swIAV vaccines could aid in reducing the amount of swIAV circulating in pig populations, thereby increasing animal welfare, limiting economic losses and lowering the risk of zoonotic swIAV transmission.
Collapse
Affiliation(s)
- Annika Graaf
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany.
| | - Philipp P Petric
- Institute of Virology, Medical Center, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Julia Sehl-Ewert
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Dinah Henritzi
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Jacqueline King
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Anne Pohlmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | | | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center, University of Freiburg, 79104 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| |
Collapse
|
32
|
Nogales A, DeDiego ML, Martínez-Sobrido L. Live attenuated influenza A virus vaccines with modified NS1 proteins for veterinary use. Front Cell Infect Microbiol 2022; 12:954811. [PMID: 35937688 PMCID: PMC9354547 DOI: 10.3389/fcimb.2022.954811] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Influenza A viruses (IAV) spread rapidly and can infect a broad range of avian or mammalian species, having a tremendous impact in human and animal health and the global economy. IAV have evolved to develop efficient mechanisms to counteract innate immune responses, the first host mechanism that restricts IAV infection and replication. One key player in this fight against host-induced innate immune responses is the IAV non-structural 1 (NS1) protein that modulates antiviral responses and virus pathogenicity during infection. In the last decades, the implementation of reverse genetics approaches has allowed to modify the viral genome to design recombinant IAV, providing researchers a powerful platform to develop effective vaccine strategies. Among them, different levels of truncation or deletion of the NS1 protein of multiple IAV strains has resulted in attenuated viruses able to induce robust innate and adaptive immune responses, and high levels of protection against wild-type (WT) forms of IAV in multiple animal species and humans. Moreover, this strategy allows the development of novel assays to distinguish between vaccinated and/or infected animals, also known as Differentiating Infected from Vaccinated Animals (DIVA) strategy. In this review, we briefly discuss the potential of NS1 deficient or truncated IAV as safe, immunogenic and protective live-attenuated influenza vaccines (LAIV) to prevent disease caused by this important animal and human pathogen.
Collapse
Affiliation(s)
- Aitor Nogales
- Centro de Investigación en Sanidad Animal (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA, CSIC), Madrid, Spain
- *Correspondence: Aitor Nogales, ; Luis Martínez-Sobrido,
| | - Marta L. DeDiego
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Martínez-Sobrido
- Department of Disease Intervention and Prevetion, Texas Biomedical Research Institute, San Antonio, TX, United States
- *Correspondence: Aitor Nogales, ; Luis Martínez-Sobrido,
| |
Collapse
|
33
|
Li B, Su G, Xiao C, Zhang J, Li H, Sun N, Lao G, Yu Y, Ren X, Qi W, Wang X, Liao M. The PB2 co-adaptation of H10N8 avian influenza virus increases the pathogenicity to chickens and mice. Transbound Emerg Dis 2022; 69:1794-1803. [PMID: 34008327 DOI: 10.1111/tbed.14157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
Avian influenza (AI) is an important zoonotic disease, which can be transmitted across species barriers to other hosts, especially humans, posing a serious threat to the poultry industry and public health. In recent years, human cases infected with the H10N8, H9N2, and H7N9 of avian influenza viruses (AIVs) have been identified frequently as have the internal genes of H7N9 and H10N8, which are derived from H9N2 viruses. The adaptive mutation of the PB2 gene is an important way for the H10N8, H9N2, and H7N9 AIVs to spread across species to adapt to new hosts. Several well-known adaptive mutations in the PB2 gene, such as E627K, D701N, and A588V, significantly enhanced the virulence of the AIVs in mammals. However, the co-adaptation of AIVs to avian and mammalian hosts is rarely studied. In this study, we found that the mutations of PB2-I292V, PB2-R389K, PB2-A588V, PB2-T598M/V, PB2-L648V, and PB2-T676M substitutions significantly increased after 2012. In addition, in our previous studies, we found that the human-origin and avian-origin of H10N8 AIVs with very high homology also have these six mutation differences in PB2 gene, and the avian-origin H10N8 strain known as JX102 with all the key amino acids on the PB2 protein in the pre-evolutionary stage, so JX102 was chosen as a model strain. Among them, PB2-A588V significantly enhanced the activity of polymerase in avian and mammalian cells. Notably, animal experiments showed that PB2-A588V substitution increased the pathogenicity and transmissibility in chickens and the virulence of mice. The combined mutations of PB2-F6 (including PB2-I292V, PB2-R389K, PB2-A588V, PB2-T598M, PB2-L648V, and PB2-T676M) obtained higher adaptability of AIVs in avians and mammals than that of the single mutation of PB2-A588V, which suggested that the PB2 588 site is a key co-adaptation site and that synergies with other mutation sites can further enhance this co-adaptability. The results of this study show that the emergence of co-adaptation not only increases the threat to avians and mammals but may also contribute to a pandemic among avians and cross the interspecies barrier to mammals.
Collapse
Affiliation(s)
- Bo Li
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- Key Laboratory of Zoonoses, Ministry of Agricultural and Rural Affairs of the People's Republic of China, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, National Development and Reform Commission of the People's Republic of China, Guangzhou, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Guanming Su
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chencheng Xiao
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiahao Zhang
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- Key Laboratory of Zoonoses, Ministry of Agricultural and Rural Affairs of the People's Republic of China, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, National Development and Reform Commission of the People's Republic of China, Guangzhou, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Huanan Li
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- Key Laboratory of Zoonoses, Ministry of Agricultural and Rural Affairs of the People's Republic of China, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, National Development and Reform Commission of the People's Republic of China, Guangzhou, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Na Sun
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guangjie Lao
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuandi Yu
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xingxing Ren
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenbao Qi
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- Key Laboratory of Zoonoses, Ministry of Agricultural and Rural Affairs of the People's Republic of China, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, National Development and Reform Commission of the People's Republic of China, Guangzhou, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| | - Xiuhui Wang
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Hebei Province Poultry Engineering Technology Research Center, Hebei University of Engineering, Handan, People's Republic of China
| | - Ming Liao
- National Avian Influenza Para-Reference Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- Key Laboratory of Zoonoses, Ministry of Agricultural and Rural Affairs of the People's Republic of China, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, National Development and Reform Commission of the People's Republic of China, Guangzhou, China
- Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, China
| |
Collapse
|
34
|
Hennig C, Graaf A, Petric PP, Graf L, Schwemmle M, Beer M, Harder T. Are pigs overestimated as a source of zoonotic influenza viruses? Porcine Health Manag 2022; 8:30. [PMID: 35773676 PMCID: PMC9244577 DOI: 10.1186/s40813-022-00274-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background Swine influenza caused by influenza A viruses (IAV) directly affects respiratory health and indirectly impairs reproduction rates in pigs causing production losses. In Europe, and elsewhere, production systems have intensified featuring fewer holdings but, in turn, increased breeding herd and litter sizes. This seems to foster swine IAV (swIAV) infections with respect to the entrenchment within and spread between holdings. Disease management of swine influenza is difficult and relies on biosecurity and vaccination measures. Recently discovered and widely proliferating forms of self-sustaining modes of swIAV infections in large swine holdings challenge these preventive concepts by generating vaccine-escape mutants in rolling circles of infection. Main body The most recent human IAV pandemic of 2009 rooted at least partly in IAV of porcine origin highlighting the zoonotic potential of swIAV. Pigs constitute a mixing vessel of IAV from different species including avian and human hosts. However, other host species such as turkey and quail but also humans themselves may also act in this way; thus, pigs are not essentially required for the generation of IAV reassortants with a multispecies origin. Since 1918, all human pandemic influenza viruses except the H2N2 virus of 1958 have been transmitted in a reverse zoonotic mode from human into swine populations. Swine populations act as long-term reservoirs of these viruses. Human-derived IAV constitute a major driver of swIAV epidemiology in pigs. Swine-to-human IAV transmissions occurred rarely and mainly sporadically as compared to avian-to-human spill-over events of avian IAV. Yet, new swIAV variants that harbor zoonotic components continue to be detected. This increases the risk that such components might eventually reassort into viruses with pandemic potential. Conclusions Domestic pig populations should not be globally stigmatized as the only or most important reservoir of potentially zoonotic IAV. The likely emergence from swine of the most recent human IAV pandemic in 2009, however, emphasized the principal risks of swine populations in which IAV circulate unimpededly. Implementation of regular and close-meshed IAV surveillance of domestic swine populations to follow the dynamics of swIAV evolution is clearly demanded. Improved algorithms for directly inferring zoonotic potential from whole IAV genome sequences as well as improved vaccines are still being sought.
Collapse
Affiliation(s)
- Christin Hennig
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Annika Graaf
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Philipp P Petric
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Laura Graf
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493, Greifswald-Insel Riems, Germany.
| |
Collapse
|
35
|
Siddoway AC, Verhoeven D, Ross KA, Wannemuehler MJ, Mallapragada SK, Narasimhan B. Structural Stability and Antigenicity of Universal Equine H3N8 Hemagglutinin Trimer upon Release from Polyanhydride Nanoparticles and Pentablock Copolymer Hydrogels. ACS Biomater Sci Eng 2022; 8:2500-2507. [PMID: 35604784 DOI: 10.1021/acsbiomaterials.2c00219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Seasonal influenza A virus infections present substantial costs to both health and economic resources each year. Current seasonal influenza vaccines provide suboptimal protection and require annual reformulation to match circulating strains. In this work, a recombinant equine H3N8 hemagglutinin trimer (rH33) known to generate cross-protective antibodies and protect animals against sublethal, heterologous virus challenge was used as a candidate vaccine antigen. Nanoadjuvants such as polyanhydride nanoparticles and pentablock copolymer hydrogels have been shown to be effective adjuvants, inducing both rapid and long-lived protective immunity against influenza A virus. In this work, polyanhydride nanoparticles and pentablock copolymer hydrogels were used to provide sustained release of the novel rH33 while also facilitating the retention of its structure and antigenicity. These studies lay the groundwork for the development of a novel universal influenza A virus nanovaccine by combining the equine H3N8 rH33 and polymeric nanoadjuvant platforms.
Collapse
Affiliation(s)
- Alaric C Siddoway
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - David Verhoeven
- Department of Veterinary Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| | | | - Michael J Wannemuehler
- Department of Veterinary Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| | - Surya K Mallapragada
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| |
Collapse
|
36
|
Shin DL, Siebert U, Haas L, Valentin-Weigand P, Herrler G, Wu NH. Primary harbor seal (Phoca vitulina) airway epithelial cells show high susceptibility to infection by a seal-derived influenza A virus (H5N8). Transbound Emerg Dis 2022; 69:e2378-e2388. [PMID: 35504691 DOI: 10.1111/tbed.14580] [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: 03/02/2022] [Revised: 04/24/2022] [Accepted: 04/29/2022] [Indexed: 11/29/2022]
Abstract
Highly pathogenic avian influenza viruses of the H5N8 subtype have been circulating in Europe and Asia since 2016, causing huge economic losses to the poultry industry. A new wave of H5Nx infections has begun in 2020. The viruses mainly infect wild birds and waterfowl; from there they spread to poultry and cause disease. Previous studies have shown that the H5N8 viruses have seldom spread to mammals; however, reports in early 2021 indicate that humans may be infected, and some incident reports indicate that H5Nx clade 2.3.4.4B virus may be transmitted to wild mammals, such as red foxes and seals. In order to get more information on how the H5N8 virus affects seals and other marine animals, here, we used primary cultures to analyze the cell tropism of the H5N8 virus, which was isolated from an infected gray seal (H5N8/Seal-2016). Primary tracheal epithelial cells were readily infected by H5N8/Seal -2016 virus; in contrast, the commonly used primary seal kidney cells required the presence of exogenous trypsin to initiate virus infection. When applied to an ex vivo precision-cut lung slice model, compared with recombinant human H3N2 virus or H9N2 LPAI virus, the H5N8/Seal-2016 virus replicated to a high titer and caused a strong detrimental effect; with these characteristics, the virus was superior to a human H3N2 virus and to an H9N2 LPAI virus. By using well-differentiated air-liquid interface cultures, we have observed that ALI cultures of canines, ferrets, and harbor seals are more sensitive to H5N8/Seal-2016 virus than are human or porcine ALI cultures, which cannot be fully explained by sialic acid distribution. Our results indicate that the airway epithelium of carnivores may be the main target of H5N8 viruses. Consideration should be given to an increased monitoring of the distribution of highly pathogenic avian influenza viruses in wild animals. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Dai-Lun Shin
- Institute of Virology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses, Hannover, Germany
| | - Ursula Siebert
- Institute for Terrestrial and Aquatic Wildlife Research, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ludwig Haas
- Institute of Virology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Peter Valentin-Weigand
- Institute of Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Georg Herrler
- Institute of Virology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nai-Huei Wu
- Department of Veterinary Medicine, National Taiwan University, Taiwan
| |
Collapse
|
37
|
Inhibition of avian influenza virus H9N2 infection by antiviral hexapeptides that target viral attachment to epithelial cells. Virus Res 2022; 313:198745. [DOI: 10.1016/j.virusres.2022.198745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/10/2022] [Accepted: 03/10/2022] [Indexed: 01/04/2023]
|
38
|
Ahmad S, Koh KY, Lee JI, Suh GH, Lee CM. Interpolation of Point Prevalence Rate of the Highly Pathogenic Avian Influenza Subtype H5N8 Second Phase Epidemic in South Korea. Vet Sci 2022; 9:vetsci9030139. [PMID: 35324867 PMCID: PMC8954420 DOI: 10.3390/vetsci9030139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022] Open
Abstract
Humans and animals are both susceptible to highly pathogenic avian influenza (HPAI) viruses. In the future, HPAI has the potential to be a source of zoonoses and pandemic disease drivers. It is necessary to identify areas of high risk that are more vulnerable to HPAI infections. In this study, we applied unbiased predictions based on known information to find points of localities with a high probability of point prevalence rate. To carry out such predictions, we utilized the inverse distance weighting (IDW) and kriging method, with the help of the R statistical computing program. The provinces of Jeollanam-do, Gyeonggi-do, Chungcheongbuk-do and Ulsan have high anticipated risk. This research might aid in the management of avian influenza threats associated with various potential risks.
Collapse
Affiliation(s)
- Saleem Ahmad
- Veterinary Public Health Lab, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea; (S.A.); (K.-Y.K.); (J.-i.L.)
| | - Kye-Young Koh
- Veterinary Public Health Lab, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea; (S.A.); (K.-Y.K.); (J.-i.L.)
| | - Jae-il Lee
- Veterinary Public Health Lab, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea; (S.A.); (K.-Y.K.); (J.-i.L.)
| | - Guk-Hyun Suh
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea;
| | - Chang-Min Lee
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea;
- Correspondence:
| |
Collapse
|
39
|
Zhang D, Yang Y, Li M, Lu Y, Liu Y, Jiang J, Liu R, Liu J, Huang X, Li G, Qu J. Ecological Barrier Deterioration Driven by Human Activities Poses Fatal Threats to Public Health due to Emerging Infectious Diseases. ENGINEERING (BEIJING, CHINA) 2022; 10:155-166. [PMID: 33903827 PMCID: PMC8060651 DOI: 10.1016/j.eng.2020.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 05/24/2023]
Abstract
The coronavirus disease 2019 (COVID-19) and concerns about several other pandemics in the 21st century have attracted extensive global attention. These emerging infectious diseases threaten global public health and raise urgent studies on unraveling the underlying mechanisms of their transmission from animals to humans. Although numerous works have intensively discussed the cross-species and endemic barriers to the occurrence and spread of emerging infectious diseases, both types of barriers play synergistic roles in wildlife habitats. Thus far, there is still a lack of a complete understanding of viral diffusion, migration, and transmission in ecosystems from a macro perspective. In this review, we conceptualize the ecological barrier that represents the combined effects of cross-species and endemic barriers for either the natural or intermediate hosts of viruses. We comprehensively discuss the key influential factors affecting the ecological barrier against viral transmission from virus hosts in their natural habitats into human society, including transmission routes, contact probability, contact frequency, and viral characteristics. Considering the significant impacts of human activities and global industrialization on the strength of the ecological barrier, ecological barrier deterioration driven by human activities is critically analyzed for potential mechanisms. Global climate change can trigger and expand the range of emerging infectious diseases, and human disturbances promote higher contact frequency and greater transmission possibility. In addition, globalization drives more transmission routes and produces new high-risk regions in city areas. This review aims to provide a new concept for and comprehensive evidence of the ecological barrier blocking the transmission and spread of emerging infectious diseases. It also offers new insights into potential strategies to protect the ecological barrier and reduce the wide-ranging risks of emerging infectious diseases to public health.
Collapse
Affiliation(s)
- Dayi Zhang
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Yunfeng Yang
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Miao Li
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Yun Lu
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Yi Liu
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Jingkun Jiang
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Ruiping Liu
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Jianguo Liu
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Xia Huang
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Guanghe Li
- School of Environment, Tsinghua University, Beijing 100084, China
| | - Jiuhui Qu
- School of Environment, Tsinghua University, Beijing 100084, China
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| |
Collapse
|
40
|
Multiple Neuraminidase Containing Influenza Virus-Like Particle Vaccines Protect Mice from Avian and Human Influenza Virus Infection. Viruses 2022; 14:v14020429. [PMID: 35216022 PMCID: PMC8875606 DOI: 10.3390/v14020429] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/04/2022] Open
Abstract
Avian influenza virus remains a threat for humans, and vaccines preventing both avian and human influenza virus infections are needed. Since virus-like particles (VLPs) expressing single neuraminidase (NA) subtype elicited limited heterosubtypic protection, VLPs expressing multiple NA subtypes would enhance the extent of heterosubtypic immunity. Here, we generated avian influenza VLP vaccines displaying H5 hemagglutinin (HA) antigen with or without avian NA subtypes (N1, N6, N8) in different combinations. BALB/c mice were intramuscularly immunized with the VLPs to evaluate the resulting homologous and heterosubtypic immunity upon challenge infections with the avian and human influenza viruses (A/H5N1, A/H3N2, A/H1N1). VLPs expressing H5 alone conferred homologous protection but not heterosubtypic protection, whereas VLPs co-expressing H5 and NA subtypes elicited both homologous and heterosubtypic protection against human influenza viruses in mice. We observed that VLP induced neuraminidase inhibitory activities (NAI), virus-neutralizing activity, and virus-specific antibody (IgG, IgA) responses were strongly correlated with the number of different NA subtype expressions on the VLPs. VLPs expressing all 3 NA subtypes resulted in the highest protection, indicated by the lowest lung titer, negligible body weight changes, and survival in immunized mice. These results suggest that expressing multiple neuraminidases in avian HA VLPs is a promising approach for developing a universal influenza A vaccine against avian and human influenza virus infections.
Collapse
|
41
|
Abstract
Globally swine influenza is one of the most important diseases of the pig industry, with various subtypes of swine influenza virus co-circulating in the field. Swine influenza can not only cause large economic losses for the pig industry but can also lead to epidemics or pandemics in the human population. We provide an overview of the pathogenic characteristics of the disease, diagnosis, risk factors for the occurrence on pig farms, impact on pigs and humans and methods to control it. This review is designed to promote understanding of the epidemiology of swine influenza which will benefit the control of the disease in both pigs and humans.
Collapse
Affiliation(s)
- Yin Li
- School of Veterinary Medicine, Murdoch University, Perth, WA Australia.,Commonwealth Scientific and Industrial Research Organisation, St. Lucia, QLD Australia
| | - Ian Robertson
- School of Veterinary Medicine, Murdoch University, Perth, WA Australia.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 China.,Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, 430070 China
| |
Collapse
|
42
|
Neuraminidase in Virus-like Particles Contributes to the Protection against High Dose of Avian Influenza Virus Challenge Infection. Pathogens 2021; 10:pathogens10101291. [PMID: 34684240 PMCID: PMC8537550 DOI: 10.3390/pathogens10101291] [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: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 11/22/2022] Open
Abstract
Neuraminidase is an important target for influenza vaccination. In this study, we generated avian influenza VLPs, expressing hemagglutinin (HA), neuraminidase (NA), HA and NA co-expressed (HANA), to evaluate the protective role of NA against a high (10LD50) and low (2LD50) dose of avian influenza virus challenge infections. A single immunization with HANA VLPs elicited the highest level of virus-specific IgG, IgG1, and IgG2a responses from the sera post-vaccination and the lungs post-challenge-infection. Potent antibody-secreting cell responses were observed from the spleens and lungs of HANA-VLP-immunized mice post-challenge-infection. HANA VLPs induced the highest CD4+ T cell, CD8+ T cell, and germinal center B cells, while strongly limiting inflammatory cytokine production in the lungs compared to other VLP immunization groups. In correlation with these findings, the lowest bodyweight losses and lung virus titers were observed from HANA VLP immunization, and all of the immunized mice survived irrespective of the challenge dose. Contrastingly, VLPs expressing either HA or NA alone failed to elicit complete protection. These results indicated that NA in VLPs played a critical role in inducing protection against a high dose of the challenge infection.
Collapse
|
43
|
Du W, de Vries E, van Kuppeveld FJM, Matrosovich M, de Haan CAM. Second sialic acid-binding site of influenza A virus neuraminidase: binding receptors for efficient release. FEBS J 2021; 288:5598-5612. [PMID: 33314755 PMCID: PMC8518505 DOI: 10.1111/febs.15668] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
Influenza A viruses (IAVs) are a major cause of human respiratory tract infections and cause significant disease and mortality. Human IAVs originate from animal viruses that breached the host species barrier. IAV particles contain sialoglycan receptor-binding hemagglutinin (HA) and receptor-destroying neuraminidase (NA) in their envelope. When IAV crosses the species barrier, the functional balance between HA and NA needs to be adjusted to the sialoglycan repertoire of the novel host species. Relatively little is known about the role of NA in host adaptation in contrast to the extensively studied HA. NA prevents virion aggregation and facilitates release of (newly assembled) virions from cell surfaces and from decoy receptors abundantly present in mucus and cell glycocalyx. In addition to a highly conserved catalytic site, NA carries a second sialic acid-binding site (2SBS). The 2SBS preferentially binds α2,3-linked sialic acids and enhances activity of the neighboring catalytic site by bringing/keeping multivalent substrates in close contact with this site. In this way, the 2SBS contributes to the HA-NA balance of virus particles and affects virus replication. The 2SBS is highly conserved in all NA subtypes of avian IAVs, with some notable exceptions associated with changes in the receptor-binding specificity of HA and host tropism. Conservation of the 2SBS is invariably lost in human (pandemic) viruses and in several other viruses adapted to mammalian host species. Preservation or loss of the 2SBS is likely to be an important factor of the viral host range.
Collapse
Affiliation(s)
- Wenjuan Du
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| | - Erik de Vries
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| | - Frank J. M. van Kuppeveld
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| | | | - Cornelis A. M. de Haan
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| |
Collapse
|
44
|
Virus-Like Particle Vaccines Against Respiratory Viruses and Protozoan Parasites. Curr Top Microbiol Immunol 2021; 433:77-106. [PMID: 33650036 DOI: 10.1007/82_2021_232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The field of vaccinology underwent massive advances over the past decades with the introduction of virus-like particles (VLPs), a supra-molecular nanoparticle vaccine platform that resembles viral structures without the ability to replicate in hosts. This innovative approach has been remarkably effective, as evidenced by its profound immunogenicity and safety. These highly desirable intrinsic properties enabled their further development as vaccines against a multitude of diseases. To date, several VLP-based vaccines have already been commercialized and many more are undergoing clinical evaluation prior to FDA approval. However, efficacious vaccines against a plethora of pathogens are still lacking, which imposes a tremendous socioeconomic burden and continues to threaten public health throughout the globe. This is especially the case for several respiratory pathogens and protozoan parasites. In this review, we briefly describe the fundamentals of VLP vaccines and the unique properties that enable these to be such valuable vaccine candidates and summarize current advances in VLP-based vaccines targeting respiratory and parasitic diseases of global importance.
Collapse
|
45
|
Fellahi S, Nassik S, Maaroufi I, Tligui NS, Touzani CD, Rawi T, Delvecchio A, Ducatez MF, Houadfi ME. Pathogenesis of Avian Influenza Virus Subtype H9N2 in Turkeys and Evaluation of Inactivated Vaccine Efficacy. Avian Dis 2021; 65:46-51. [PMID: 34339121 DOI: 10.1637/aviandiseases-d-20-00067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/20/2020] [Indexed: 11/05/2022]
Abstract
Avian influenza H9N2 viruses circulate in all types of poultry species, including turkeys, and cause significant losses for the poultry industry in many parts of the word. The aim of this study was to assess the pathogenesis of the Moroccan avian influenza virus (AIV) H9N2 under experimental conditions in turkeys and the protection efficacy of an inactivated commercial vaccine against AIV H9N2. Unvaccinated turkeys showed marked depression sinusitis, respiratory distress characterized by bronchiolar and tracheal rales of moderate severity, and a mortality rate of 50%. Postmortem examinations of dead and euthanatized birds revealed the presence of fibrinous tracheitis and airsacculitis lesions. Vaccination reduced the mortality rate to 20%. Vaccinated birds recovered at day 10 postchallenge, and only 12.5% (1/8) and 37.5% of birds still displayed fibrinous and nonfibrinous airsacculitis lesions, respectively, at day 15 postinoculation. Viral shedding in cloacal and tracheal swabs was lower in vaccinated than in control birds. Although viral RNA was detected in the cloacal swabs of all unvaccinated turkeys at day 3 postinoculation, only 50% of the vaccinated turkeys were positive for virus detection. At day 11 postinoculation, no viral RNA was detected in oropharyngeal swabs of vaccinated turkeys, whereas 40% of the unvaccinated turkeys were still shedding virus.
Collapse
Affiliation(s)
- Siham Fellahi
- Unité de Pathologie Aviaire, Département de Pathologie et Santé Publique Vétérinaire, Institut Agronomique et Vétérinaire Hassan II, Rabat, Morocco, ,
| | - Saadia Nassik
- Unité de Pathologie Aviaire, Département de Pathologie et Santé Publique Vétérinaire, Institut Agronomique et Vétérinaire Hassan II, Rabat, Morocco
| | - Imane Maaroufi
- Unité de Pathologie Aviaire, Département de Pathologie et Santé Publique Vétérinaire, Institut Agronomique et Vétérinaire Hassan II, Rabat, Morocco
| | - Nour-Said Tligui
- Unité de Pathologie Aviaire, Département de Pathologie et Santé Publique Vétérinaire, Institut Agronomique et Vétérinaire Hassan II, Rabat, Morocco
| | - Charifa Drissi Touzani
- Unité de Pathologie Aviaire, Département de Pathologie et Santé Publique Vétérinaire, Institut Agronomique et Vétérinaire Hassan II, Rabat, Morocco
| | | | | | | | - Mohamed El Houadfi
- Unité de Pathologie Aviaire, Département de Pathologie et Santé Publique Vétérinaire, Institut Agronomique et Vétérinaire Hassan II, Rabat, Morocco
| |
Collapse
|
46
|
Temporal Dynamics of Influenza A(H5N1) Subtype before and after the Emergence of H5N8. Viruses 2021; 13:v13081565. [PMID: 34452430 PMCID: PMC8412109 DOI: 10.3390/v13081565] [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: 06/28/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 01/10/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) viruses continue to circulate worldwide, causing numerous outbreaks among bird species and severe public health concerns. H5N1 and H5N8 are the two most fundamental HPAI subtypes detected in birds in the last two decades. The two viruses may compete with each other while sharing the same host population and, thus, suppress the spread of one of the viruses. In this study, we performed a statistical analysis to investigate the temporal correlation of the HPAI H5N1 and HPAI H5N8 subtypes using globally reported data in 2015-2020. This was joined with an in-depth analysis using data generated via our national surveillance program in Egypt. A total of 6412 outbreaks were reported worldwide during this period, with 39% (2529) as H5N1 and 61% (3883) as H5N8. In Egypt, 65% of positive cases were found in backyards, while only 12% were found in farms and 23% in live bird markets. Overall, our findings depict a trade-off between the number of positive H5N1 and H5N8 samples around early 2017, which is suggestive of the potential replacement between the two subtypes. Further research is still required to elucidate the underpinning mechanisms of this competitive dynamic. This, in turn, will implicate the design of effective strategies for disease control.
Collapse
|
47
|
Epidemiology, Genetic Characterization, and Pathogenesis of Avian Influenza H5N8 Viruses Circulating in Northern and Southern Parts of Egypt, 2017-2019. Animals (Basel) 2021; 11:ani11082208. [PMID: 34438666 PMCID: PMC8388380 DOI: 10.3390/ani11082208] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary During 2020–2021, highly pathogenic avian influenza (HPAI) viruses of subtype H5N8 were spreading rapidly, and two genetically distinct lineages were detected in Europe, the Middle East, and Southeast Asia. HPAI H5N8 viruses have been circulating in Egyptian poultry flocks since 2016. In this study, 74 commercial chicken farms tested positive for HPAI H5N8 virus. Genetic characterization of the hemagglutinin (HA) and the neuraminidase (NA) of Egyptian HPAI H5N8 viruses showed a relationship with those recently isolated in Europe. Abstract Highly pathogenic avian influenza (HPAI) viruses of subtype H5N8 continue to circulate, causing huge economic losses and serious impact on poultry production worldwide. Recently, HPAIV H5N8 has been spreading rapidly, and a large number of HPAI H5N8 outbreaks have been reported in Eurasia 2020–2021. In this study, we conducted an epidemiological survey of HPAI H5N8 virus at different geographical locations in Egypt from 2017 to 2019. This was followed by genetic and pathogenic studies. Our findings highlight the wide spread of HPAI H5N8 viruses in Egypt, including in 22 governorates. The genetic analyses of the hemagglutinin (HA) and neuraminidase (NA) gene segments emphasized a phylogenetic relatedness between the Egyptian HPAI H5N8 viruses and viruses of clade 2.3.4.4b recently isolated in Europe. These findings suggest that a potential back transmission of Egyptian HPAI H5N8 virus has occurred from domestic poultry in Egypt to migratory wild birds, followed by further spread to different countries. This highlights the importance of continuous epidemiological and genetic studies of AIVs at the domestic–wild bird interface.
Collapse
|
48
|
Delgado-Hernández B, Mugica L, Acosta M, Pérez F, Montano DDLN, Abreu Y, Ayala J, Percedo MI, Alfonso P. Knowledge, Attitudes, and Risk Perception Toward Avian Influenza Virus Exposure Among Cuban Hunters. Front Public Health 2021; 9:644786. [PMID: 34368040 PMCID: PMC8342762 DOI: 10.3389/fpubh.2021.644786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/28/2021] [Indexed: 11/24/2022] Open
Abstract
A critical step for decreasing zoonotic disease threats is to have a good understanding of the associated risks. Hunters frequently handle potentially infected birds, so they are more at risk of being exposed to zoonotic avian pathogens, including avian influenza viruses (AIVs). The objective of the current study was to gain a better understanding of Cuban hunters' general hunting practices, focusing on their knowledge and risk perception on avian influenza. An anonymous and voluntary semi-structured questionnaire was designed and applied to 398 hunters. Multiple correspondence analyses found relationships with potential exposure of AIVs to people and domestic animals. The main associated risks factors identified were not taking the annual flu vaccine (60.1%) and not cleaning hunting knives (26.3%); Direct contact with water (32.1%), cleaning wild birds at home (33.2%); receiving assistance during bird cleaning (41.9%), keeping poultry at home (56.5%) and feeding domestic animals with wild bird leftovers (30.3%) were also identified as significant risk factors. The lack of use of some protective measures reported by hunters had no relationship with their awareness on avian influenza, which may imply a lack of such knowledge. The results evidenced that more effective risk communication strategies about the consequences of AIVs infecting human or other animals, and the importance of reducing such risks, are urgently needed.
Collapse
Affiliation(s)
- Beatriz Delgado-Hernández
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba
| | - Lourdes Mugica
- Bird Ecology Group, Biology Faculty, Havana University, Vedado, Cuba
| | - Martin Acosta
- Bird Ecology Group, Biology Faculty, Havana University, Vedado, Cuba
| | - Frank Pérez
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba.,Department of Veterinary Medicine, Faculty of Agricultural Sciences, University of Granma, Bayamo, Cuba
| | - Damarys de Las Nieves Montano
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba
| | - Yandy Abreu
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba
| | - Joel Ayala
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba
| | - María Irian Percedo
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba
| | - Pastor Alfonso
- Epidemiology Group, National Center for Animal and Plant Health (CENSA), World Organisation for Animal Health (OIE) Collaborating Center for the Reduction of the Risk of Disaster in Animal Health, San José de las Lajas, Cuba
| |
Collapse
|
49
|
Kim WH, Cho S. Estimation of the Basic Reproduction Numbers of the Subtypes H5N1, H5N8, and H5N6 During the Highly Pathogenic Avian Influenza Epidemic Spread Between Farms. Front Vet Sci 2021; 8:597630. [PMID: 34250054 PMCID: PMC8264784 DOI: 10.3389/fvets.2021.597630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
It is important to understand pathogen transmissibility in a population to establish an effective disease prevention policy. The basic reproduction number (R 0) is an epidemiologic parameter for understanding the characterization of disease and its dynamics in a population. We aimed to estimate the R 0 of the highly pathogenic avian influenza (HPAI) subtypes H5N1, H5N8, and H5N6, which were associated with nine outbreaks in Korea between 2003 and 2018, to understand the epidemic transmission of each subtype. According to HPAI outbreak reports of the Animal and Plant Quarantine Agency, we estimated the generation time by calculating the time of infection between confirmed HPAI-positive farms. We constructed exponential growth and maximum likelihood (ML) models to estimate the basic reproduction number, which assumes the number of secondary cases infected by the index case. The Kruskal-Wallis test was used to analyze the epidemic statistics between subtypes. The estimated generation time of H5N1, H5N8, and H5N6 were 4.80 days [95% confidence interval (CI) 4.23-5.38] days, 7.58 (95% CI 6.63-8.46), and 5.09 days (95% CI 4.44-5.74), respectively. A pairwise comparison showed that the generation time of H5N8 was significantly longer than that of the subtype H5N1 (P = 0.04). Based on the ML model, R 0 was estimated as 1.69 (95% CI 1.48-2.39) for subtype H5N1, 1.60 (95%CI 0.97-2.23) for subtype H5N8, and 1.49 (95%CI 0.94-2.04) for subtype H5N6. We concluded that R 0 estimates may be associated with the poultry product system, climate, species specificity based on the HPAI virus subtype, and prevention policy. This study provides an insight on the transmission and dynamics patterns of various subtypes of HPAI occurring worldwide. Furthermore, the results are useful as scientific evidence for establishing a disease control policy.
Collapse
Affiliation(s)
| | - Seongbeom Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
50
|
Tong ZWM, Karawita AC, Kern C, Zhou H, Sinclair JE, Yan L, Chew KY, Lowther S, Trinidad L, Challagulla A, Schat KA, Baker ML, Short KR. Primary Chicken and Duck Endothelial Cells Display a Differential Response to Infection with Highly Pathogenic Avian Influenza Virus. Genes (Basel) 2021; 12:genes12060901. [PMID: 34200798 PMCID: PMC8230508 DOI: 10.3390/genes12060901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 01/12/2023] Open
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) in gallinaceous poultry are associated with viral infection of the endothelium, the induction of a ‘cytokine storm, and severe disease. In contrast, in Pekin ducks, HPAIVs are rarely endothelial tropic, and a cytokine storm is not observed. To date, understanding these species-dependent differences in pathogenesis has been hampered by the absence of a pure culture of duck and chicken endothelial cells. Here, we use our recently established in vitro cultures of duck and chicken aortic endothelial cells to investigate species-dependent differences in the response of endothelial cells to HPAIV H5N1 infection. We demonstrate that chicken and duck endothelial cells display a different transcriptional response to HPAI H5N1 infection in vitro—with chickens displaying a more pro-inflammatory response to infection. As similar observations were recorded following in vitro stimulation with the viral mimetic polyI:C, these findings were not specific to an HPAIV H5N1 infection. However, similar species-dependent differences in the transcriptional response to polyI:C were not observed in avian fibroblasts. Taken together, these data demonstrate that chicken and duck endothelial cells display a different response to HPAIV H5N1 infection, and this may help account for the species-dependent differences observed in inflammation in vivo.
Collapse
Affiliation(s)
- Zhen Wei Marcus Tong
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; (Z.W.M.T.); (A.C.K.); (J.E.S.); (L.Y.); (K.Y.C.)
| | - Anjana C. Karawita
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; (Z.W.M.T.); (A.C.K.); (J.E.S.); (L.Y.); (K.Y.C.)
- CSIRO, Australian Centre for Disease Preparedness, Health, and Biosecurity Business Unit, Geelong 3219, Australia; (S.L.); (L.T.); (A.C.); (M.L.B.)
| | - Colin Kern
- Department of Animal Science, University of California, Davis, CA 95616, USA; (C.K.); (H.Z.)
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, CA 95616, USA; (C.K.); (H.Z.)
| | - Jane E. Sinclair
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; (Z.W.M.T.); (A.C.K.); (J.E.S.); (L.Y.); (K.Y.C.)
| | - Limin Yan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; (Z.W.M.T.); (A.C.K.); (J.E.S.); (L.Y.); (K.Y.C.)
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; (Z.W.M.T.); (A.C.K.); (J.E.S.); (L.Y.); (K.Y.C.)
| | - Sue Lowther
- CSIRO, Australian Centre for Disease Preparedness, Health, and Biosecurity Business Unit, Geelong 3219, Australia; (S.L.); (L.T.); (A.C.); (M.L.B.)
| | - Lee Trinidad
- CSIRO, Australian Centre for Disease Preparedness, Health, and Biosecurity Business Unit, Geelong 3219, Australia; (S.L.); (L.T.); (A.C.); (M.L.B.)
| | - Arjun Challagulla
- CSIRO, Australian Centre for Disease Preparedness, Health, and Biosecurity Business Unit, Geelong 3219, Australia; (S.L.); (L.T.); (A.C.); (M.L.B.)
| | - Karel A. Schat
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA;
| | - Michelle L. Baker
- CSIRO, Australian Centre for Disease Preparedness, Health, and Biosecurity Business Unit, Geelong 3219, Australia; (S.L.); (L.T.); (A.C.); (M.L.B.)
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; (Z.W.M.T.); (A.C.K.); (J.E.S.); (L.Y.); (K.Y.C.)
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane 4072, Australia
- Correspondence:
| |
Collapse
|