1
|
De Meyer A, Meuleman P. Preclinical animal models to evaluate therapeutic antiviral antibodies. Antiviral Res 2024; 225:105843. [PMID: 38548022 DOI: 10.1016/j.antiviral.2024.105843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 04/05/2024]
Abstract
Despite the availability of effective preventative vaccines and potent small-molecule antiviral drugs, effective non-toxic prophylactic and therapeutic measures are still lacking for many viruses. The use of monoclonal and polyclonal antibodies in an antiviral context could fill this gap and provide effective virus-specific medical interventions. In order to develop these therapeutic antibodies, preclinical animal models are of utmost importance. Due to the variability in viral pathogenesis, immunity and overall characteristics, the most representative animal model for human viral infection differs between virus species. Therefore, throughout the years researchers sought to find the ideal preclinical animal model for each virus. The most used animal models in preclinical research include rodents (mice, ferrets, …) and non-human primates (macaques, chimpanzee, ….). Currently, antibodies are tested for antiviral efficacy against a variety of viruses including different hepatitis viruses, human immunodeficiency virus (HIV), influenza viruses, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and rabies virus. This review provides an overview of the current knowledge about the preclinical animal models that are used for the evaluation of therapeutic antibodies for the abovementioned viruses.
Collapse
Affiliation(s)
- Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
2
|
Anjum FR, Anam S, Rahman SU, Ali S, Aslam MA, Rizvi F, Asif M, Abdullah RM, Abaidullah M, Shakir MZ, Goraya MU. Anti-chicken type I IFN countermeasures by major avian RNA viruses. Virus Res 2020; 286:198061. [PMID: 32561378 DOI: 10.1016/j.virusres.2020.198061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/27/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022]
Abstract
Chicken type I interferons (type I IFNs) are key antiviral players of the chicken innate immune system and are considered potent antiviral agents against avian viral pathogens. Chicken type I IFNs are divided into three subtypes namely, chIFN-α, chIFN-β, and chIFN-κ. Viral pathogen-associated molecular patterns (PAMPs) recognized by their corresponding specific PRRs (pattern recognition receptors) induce the expression of chicken type I IFNs. Interaction of chicken type I IFNs with their subsequent IFN receptors results in the activation of the JAK-STAT pathway, which in turn activates hundreds of chicken interferon-stimulated genes (chISGs). These chISGs establish an antiviral state in neighboring cells and prevent the replication and dissemination of viruses within chicken cells. Chicken type I IFNs activate different pathways that constitute major antiviral innate defense mechanisms in chickens. However, evolutionary mechanisms in viruses have made them resistant to these antiviral players by manipulating host innate immune pathways. This review focuses on the underlying molecular mechanisms employed by avian RNA viruses to counteract chicken type I IFNs and chISGs through different viral proteins. This may help to understand host-pathogen interactions and the development of novel therapeutic strategies to control viral infections in poultry.
Collapse
Affiliation(s)
| | - Sidra Anam
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Sajjad Ur Rahman
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Sultan Ali
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | | | - Farzana Rizvi
- Department of Pathology, Faculty of Veterinary Science, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Asif
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | | | - Muhammad Abaidullah
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | | | | |
Collapse
|
3
|
Kiseleva I, Rekstin A, Al Farroukh M, Bazhenova E, Katelnikova A, Puchkova L, Rudenko L. Non-Mouse-Adapted H1N1pdm09 Virus as a Model for Influenza Research. Viruses 2020; 12:v12060590. [PMID: 32485821 PMCID: PMC7354452 DOI: 10.3390/v12060590] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/26/2022] Open
Abstract
The number of lung-adapted influenza viruses is limited. Most of them are not antigenically related to current circulating viruses. Viruses similar to recent strains are required for screening modern antiviral compounds and studying new vaccine candidates against novel influenza viruses. The process by which an influenza virus adapts to a new host is rather difficult. The aim of this study was to select a non-adapted current virus whose major biological properties correspond to those of classical lab-adapted viruses. Mice were inoculated intranasally with non-lung-adapted influenza viruses of subtype H1N1pdm09. They were monitored closely for body weight loss, mortality outcomes and gross pathology for 14 days following inoculation, as well as viral replication in lung tissue. Lung-adapted PR8 virus was used as a control. The tested viruses multiplied equally well in the lower respiratory tract of mice without prior adaptation but dramatically differed in lethality; the differences in their toxicity and pathogenicity in mice were established. A/South Africa/3626/2013 (H1N1)pdm09 virus was found to be an appropriate candidate to replace PR8 as a model virus for influenza research. No prior adaptation to the animal model is needed to reach the pathogenicity level of the classical mouse-adapted PR8 virus.
Collapse
Affiliation(s)
- Irina Kiseleva
- Department of Virology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, 197376 St Petersburg, Russia; (A.R.); (M.A.F.); (E.B.); (L.P.); (L.R.)
- Correspondence:
| | - Andrey Rekstin
- Department of Virology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, 197376 St Petersburg, Russia; (A.R.); (M.A.F.); (E.B.); (L.P.); (L.R.)
| | - Mohammad Al Farroukh
- Department of Virology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, 197376 St Petersburg, Russia; (A.R.); (M.A.F.); (E.B.); (L.P.); (L.R.)
| | - Ekaterina Bazhenova
- Department of Virology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, 197376 St Petersburg, Russia; (A.R.); (M.A.F.); (E.B.); (L.P.); (L.R.)
| | - Anastasia Katelnikova
- Department of Toxicology and Microbiology, Institute of Preclinical Research Ltd., 188663 St Petersburg, Russia;
| | - Ludmila Puchkova
- Department of Virology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, 197376 St Petersburg, Russia; (A.R.); (M.A.F.); (E.B.); (L.P.); (L.R.)
| | - Larisa Rudenko
- Department of Virology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine”, 197376 St Petersburg, Russia; (A.R.); (M.A.F.); (E.B.); (L.P.); (L.R.)
| |
Collapse
|
4
|
Comparison of nucleic acid extraction methods for next-generation sequencing of avian influenza A virus from ferret respiratory samples. J Virol Methods 2019; 270:95-105. [DOI: 10.1016/j.jviromet.2019.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/13/2019] [Accepted: 04/15/2019] [Indexed: 11/13/2022]
|
5
|
Pulit-Penaloza JA, Belser JA, Tumpey TM, Maines TR. Sowing the Seeds of a Pandemic? Mammalian Pathogenicity and Transmissibility of H1 Variant Influenza Viruses from the Swine Reservoir. Trop Med Infect Dis 2019; 4:E41. [PMID: 30818793 PMCID: PMC6473686 DOI: 10.3390/tropicalmed4010041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/02/2019] [Accepted: 02/20/2019] [Indexed: 01/01/2023] Open
Abstract
Emergence of genetically and antigenically diverse strains of influenza to which the human population has no or limited immunity necessitates continuous risk assessments to determine the likelihood of these viruses acquiring adaptations that facilitate sustained human-to-human transmission. As the North American swine H1 virus population has diversified over the last century by means of both antigenic drift and shift, in vivo assessments to study multifactorial traits like mammalian pathogenicity and transmissibility of these emerging influenza viruses are critical. In this review, we examine genetic, molecular, and pathogenicity and transmissibility data from a panel of contemporary North American H1 subtype swine-origin viruses isolated from humans, as compared to H1N1 seasonal and pandemic viruses, including the reconstructed 1918 virus. We present side-by-side analyses of experiments performed in the mouse and ferret models using consistent experimental protocols to facilitate enhanced interpretation of in vivo data. Contextualizing these analyses in a broader context permits a greater appreciation of the role that in vivo risk assessment experiments play in pandemic preparedness. Collectively, we find that despite strain-specific heterogeneity among swine-origin H1 viruses, contemporary swine viruses isolated from humans possess many attributes shared by prior pandemic strains, warranting heightened surveillance and evaluation of these zoonotic viruses.
Collapse
Affiliation(s)
- Joanna A Pulit-Penaloza
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Terrence M Tumpey
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| |
Collapse
|
6
|
Belser JA, Maines TR, Tumpey TM. Importance of 1918 virus reconstruction to current assessments of pandemic risk. Virology 2018; 524:45-55. [PMID: 30142572 PMCID: PMC9036538 DOI: 10.1016/j.virol.2018.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/25/2018] [Accepted: 08/09/2018] [Indexed: 01/13/2023]
Abstract
Reconstruction of the 1918 influenza virus has facilitated considerable advancements in our understanding of this extraordinary pandemic virus. However, the benefits of virus reconstruction are not limited to this one strain. Here, we provide an overview of laboratory studies which have evaluated the reconstructed 1918 virus, and highlight key discoveries about determinants of virulence and transmissibility associated with this virus in mammals. We further discuss recent and current pandemic threats from avian and swine reservoirs, and provide specific examples of how reconstruction of the 1918 pandemic virus has improved our ability to contextualize research employing novel and emerging strains. As influenza viruses continue to evolve and pose a threat to human health, studying past pandemic viruses is key to future preparedness efforts.
Collapse
Affiliation(s)
- Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Terrence M Tumpey
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
7
|
Pathogenesis and Transmission of Genetically Diverse Swine-Origin H3N2 Variant Influenza A Viruses from Multiple Lineages Isolated in the United States, 2011-2016. J Virol 2018; 92:JVI.00665-18. [PMID: 29848587 DOI: 10.1128/jvi.00665-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/24/2018] [Indexed: 11/20/2022] Open
Abstract
While several swine-origin influenza A H3N2 variant (H3N2v) viruses isolated from humans prior to 2011 have been previously characterized for their virulence and transmissibility in ferrets, the recent genetic and antigenic divergence of H3N2v viruses warrants an updated assessment of their pandemic potential. Here, four contemporary H3N2v viruses isolated during 2011 to 2016 were evaluated for their replicative ability in both in vitro and in vivo in mammalian models as well as their transmissibility among ferrets. We found that all four H3N2v viruses possessed similar or enhanced replication capacities in a human bronchial epithelium cell line (Calu-3) compared to a human seasonal influenza virus, suggestive of strong fitness in human respiratory tract cells. The majority of H3N2v viruses examined in our study were mildly virulent in mice and capable of replicating in mouse lungs with different degrees of efficiency. In ferrets, all four H3N2v viruses caused moderate morbidity and exhibited comparable titers in the upper respiratory tract, but only 2 of the 4 viruses replicated in the lower respiratory tract in this model. Furthermore, despite efficient transmission among cohoused ferrets, recently isolated H3N2v viruses displayed considerable variance in their ability to transmit by respiratory droplets. The lack of a full understanding of the molecular correlates of virulence and transmission underscores the need for close genotypic and phenotypic monitoring of H3N2v viruses and the importance of continued surveillance to improve pandemic preparedness.IMPORTANCE Swine-origin influenza viruses of the H3N2 subtype, with the hemagglutinin (HA) and neuraminidase (NA) derived from historic human seasonal influenza viruses, continue to cross species barriers and cause human infections, posing an indelible threat to public health. To help us better understand the potential risk associated with swine-origin H3N2v viruses that emerged in the United States during the 2011-2016 influenza seasons, we use both in vitro and in vivo models to characterize the abilities of these viruses to replicate, cause disease, and transmit in mammalian hosts. The efficient respiratory droplet transmission exhibited by some of the H3N2v viruses in the ferret model combined with the existing evidence of low immunity against such viruses in young children and older adults highlight their pandemic potential. Extensive surveillance and risk assessment of H3N2v viruses should continue to be an essential component of our pandemic preparedness strategy.
Collapse
|
8
|
Luo K, Zhang K, Liu L, Shen X, Jiao P, Song Y, Lv J, Wang M, Liu Y, Qi W, Ren T, Irwin DM, Liao M, Shen Y. The genetic and phylogenetic analysis of a highly pathogenic influenza A H5N6 virus from a heron, southern China, 2013. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2018; 59:72-74. [PMID: 29409937 DOI: 10.1016/j.meegid.2018.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/12/2018] [Accepted: 01/29/2018] [Indexed: 02/08/2023]
Abstract
The H5N6 highly pathogenic avian influenza viruses (HPAIVs) have circulated within poultry in China since 2013. Infections of H5N6 in wild birds were reported since 2014. In order to investigate the infection history of H5N6 in wild birds, we conducted a retrospective analysis of H5 positive wild bird samples collected in 2013, the year H5N6 was discovered in poultry. We isolated a new HPAI H5N6 virus from a dead heron collected in 2013. The virus had high identity in all eight gene sequences to those collected from poultry in 2013 (for example, A/chicken/Shenzhen/1845/2013, 99.1%-99.7%). Our findings revealed that H5N6 HPAIVs infected wild birds in southern China since the emergence of H5N6 in poultry in 2013. The co-circulation of H5N6 between wild birds and poultry is very close, and should raise our attention more.
Collapse
Affiliation(s)
- Kaijian Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Kai Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Lu Liu
- Shantou University Medical College, Shantou 515041, China
| | - Xuejuan Shen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Peirong Jiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yafen Song
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jiamin Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Mei Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yongfa Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Wenbao Qi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - David M Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto M5S 1A8, Canada
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China.
| | - Yongyi Shen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China; Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
9
|
Pathogenicity and transmissibility of three avian influenza A (H5N6) viruses isolated from wild birds. J Infect 2018; 76:286-294. [PMID: 29307740 DOI: 10.1016/j.jinf.2017.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/16/2017] [Accepted: 12/20/2017] [Indexed: 11/22/2022]
Abstract
Since 2013, highly pathogenic H5N6 avian influenza viruses (AIVs) have emerged in poultry and caused sporadic human infections in Asia. The recent discovery of three new avian H5N6 viruses - A/oriental magpie-robin/Guangdong/SW8/2014 (H5N6), A/common moorhen/Guangdong/GZ174/2014 (H5N6) and A/Pallas's sandgrouse/Guangdong/ZH283/2015 (H5N6) - isolated from apparently healthy wild birds in Southern China in 2014-2015 raises great concern for the spread of these highly pathogenic AIVs (HPAIVs) and their potential threat to human and animal health. In our study, we conducted animal experiments and tested their pathogenicity in ducks, chickens and mice. Ducks can carry and shed the H5N6 HPAIVs, but show no ill effects. On the other hand, these H5N6 HPAIVs can efficiently infect, transmit and cause death in chickens. Due to the overlap of habitats, domestic ducks play an important role in circulating AIVs between poultry and wild birds. Our results raise the possibility that wild birds disseminate these H5N6 HPAIVs to poultry along their flyways and thus pose a great threat to the poultry industry. These viruses are also highly pathogenic to mice, suggesting they pose a potential threat to mammals and, thus, public health. One virus isolated in 2015 replicates much more efficiently and is more lethal in these animals than the two other viruses isolated in 2014. It seems that the H5N6 viruses tend to be more lethal as time passes. Therefore, it is necessary to vigilantly monitor H5N6 HPAIVs in wild birds and poultry.
Collapse
|
10
|
Abstract
Avian influenza A H7N9 viruses that emerged in China in 2013 have reappeared each year, causing more than 1 600 severe human infections. As these viruses have evolved in nature, they have gained some and can gain additional virulence determinants that enhance their risk for humans, underlining the urgent need to control and eradicate H7N9 viruses in China.
Collapse
|
11
|
Genomic signature analysis of the recently emerged highly pathogenic A(H5N8) avian influenza virus: implying an evolutionary trend for bird-to-human transmission. Microbes Infect 2017; 19:597-604. [PMID: 28889970 DOI: 10.1016/j.micinf.2017.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/14/2017] [Accepted: 08/18/2017] [Indexed: 11/20/2022]
Abstract
In early 2014, a novel subclade (2.3.4.4) of the highly pathogenic avian influenza (HPAI) A(H5N8) virus caused the first outbreak in domestic ducks and migratory birds in South Korea. Since then, it has spread to 44 countries and regions. To date, no human infections with A(H5N8) virus have been reported, but the possibility cannot be excluded. By analyzing the genomic signatures of A(H5N8) strains, we found that among the 47 species-associated signature positions, three positions exhibited human-like signatures (HLS), including PA-404S, PB2-613I and PB2-702R and that mutation trend of host signatures of avian A(H5N8) is different before and after 2014. About 82% of A(H5N8) isolates collected after January of 2014 carried the 3 HLS (PA-404S/PB2-613I/PB2-702R) in combination, while none of isolates collected before 2014 had this combination. Furthermore, the HA protein had S137A and S227R substitutions in the receptor-binding site and A160T in the glycosylation site, potentially increasing viral ability to bind human-type receptors. Based on these findings, the newly emerged HPAI A(H5N8) isolates show an evolutionary trend toward gaining more HLS and, along with it, the potential for bird-to-human transmissibility. Therefore, more extensive surveillance of this rapidly spreading HPAI A(H5N8) and preparedness against its potential pandemic are urgently needed.
Collapse
|
12
|
Cui J, Qu N, Guo Y, Cao L, Wu S, Mei K, Sun H, Lu Y, Qin Z, Jiao P, Liao M. Phylogeny, Pathogenicity, and Transmission of H5N1 Avian Influenza Viruses in Chickens. Front Cell Infect Microbiol 2017; 7:328. [PMID: 28770175 PMCID: PMC5515836 DOI: 10.3389/fcimb.2017.00328] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/03/2017] [Indexed: 11/13/2022] Open
Abstract
We analyzed five H5N1 avian influenza viruses (AIVs) isolated from different birds in 2012 in China. Based on whole-genome sequences, we divided the viruses into four genotypes. The DKE26, GSE43, and DKE53 viruses belonged to Genotypes 1-3, respectively. The CKE93 and CKE96 viruses were classified into Genotype 4. Genotypes 1-3 correspond to the viruses containing the HA gene of clade 2.3.2, and Genotype 4 is the virus that bears the HA gene of clade 7.2. To better understand the pathogenicity and transmission of the viruses, we infected chickens with 103 EID50/0.1 ml GSE43 (clade 2.3.2) or CKE93 (clade 7.2) virus. Our results revealed that 6 of 7 specific-pathogen-free (SPF) chickens inoculated with GSE43 virus were dead before 7-day post-infection, but all the SPF chickens inoculated with CKE93 virus survived the infection. Both the GSE43 and CKE93 viruses replicated systemically in chickens. The virus titers of GSE43 virus in tested organs were obviously higher than those of CKE93 virus. Our results revealed that the pathogenicity and replication of GSE43 in chickens was much higher than those of CKE93. The GSE43 virus could transmit between chickens, but the CKE93 could not transmit between chickens by naïve contact. Therefore, different clades of H5N1 AIVs possessed variable pathogenicities and transmission abilities among chickens. Our study contributes to knowledge of pathogenic variations of prevalent H5N1 viruses.
Collapse
Affiliation(s)
- Jin Cui
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Nannan Qu
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Yang Guo
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Lan Cao
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,Guangzhou Center for Disease Control and PreventionGuangzhou, China
| | - Siyu Wu
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Kun Mei
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Hailiang Sun
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Yiliang Lu
- Animal & Plant Inspection and Quarantine Technology Center of Shenzhen Entry-Exit Inspection and Quarantine Bureau of ChinaShenzhen, China.,Department of Biotechnology, College of Life Sciences and Oceanography, Shenzhen UniversityShenzhen, China
| | - Zhifeng Qin
- Animal & Plant Inspection and Quarantine Technology Center of Shenzhen Entry-Exit Inspection and Quarantine Bureau of ChinaShenzhen, China
| | - Peirong Jiao
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| | - Ming Liao
- Animal Infectious Diseases Laboratory, College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China.,Key Laboratory of Zoonosis, Ministry of AgricultureGuangzhou, China
| |
Collapse
|
13
|
Camp JV, Jonsson CB. A Role for Neutrophils in Viral Respiratory Disease. Front Immunol 2017; 8:550. [PMID: 28553293 PMCID: PMC5427094 DOI: 10.3389/fimmu.2017.00550] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 04/24/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are immune cells that are well known to be present during many types of lung diseases associated with acute respiratory distress syndrome (ARDS) and may contribute to acute lung injury. Neutrophils are poorly studied with respect to viral infection, and specifically to respiratory viral disease. Influenza A virus (IAV) infection is the cause of a respiratory disease that poses a significant global public health concern. Influenza disease presents as a relatively mild and self-limiting although highly pathogenic forms exist. Neutrophils increase in the respiratory tract during infection with mild seasonal IAV, moderate and severe epidemic IAV infection, and emerging highly pathogenic avian influenza (HPAI). During severe influenza pneumonia and HPAI infection, the number of neutrophils in the lower respiratory tract is correlated with disease severity. Thus, comparative analyses of the relationship between IAV infection and neutrophils provide insights into the relative contribution of host and viral factors that contribute to disease severity. Herein, we review the contribution of neutrophils to IAV disease pathogenesis and to other respiratory virus infections.
Collapse
Affiliation(s)
- Jeremy V Camp
- Institute of Virology, University of Veterinary Medicine at Vienna, Vienna, Austria
| | - Colleen B Jonsson
- Department of Microbiology, University of Tennessee-Knoxville, Knoxville, TN, USA
| |
Collapse
|
14
|
Kang Y, Liu L, Feng M, Yuan R, Huang C, Tan Y, Gao P, Xiang D, Zhao X, Li Y, Irwin DM, Shen Y, Ren T. Highly pathogenic H5N6 influenza A viruses recovered from wild birds in Guangdong, southern China, 2014-2015. Sci Rep 2017; 7:44410. [PMID: 28294126 PMCID: PMC5353559 DOI: 10.1038/srep44410] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 02/10/2017] [Indexed: 02/05/2023] Open
Abstract
Since 2013, highly pathogenic (HP) H5N6 influenza A viruses (IAVs) have emerged in poultry in Asia, especially Southeast Asia. These viruses have also caused sporadic infections in humans within the same geographic areas. Active IAV surveillance in wild birds sampled in Guangdong province, China from August 2014 through February 2015 resulted in the recovery of three H5N6 IAVs. These H5N6 IAV isolates possess the basic amino acid motif at the HA1-HA2 cleavage site that is associated with highly pathogenic IAVs infecting chickens. Noteworthy findings include: (1) the HP H5N6 IAV isolates were recovered from three species of apparently healthy wild birds (most other isolates of HP H5N6 IAV in Asia are recovered from dead wild birds or fecal samples in the environment) and (2) these isolates were apparently the first recoveries of HP H5N6 IAV for two of the three species thus expanding the demonstrated natural host range for these lineages of virus. This investigation provides additional insight into the natural history of HP H5N6 IAVs and identifies the occurrence of non-lethal, HP H5N6 IAV infections in wild birds thereby demonstrating the value of active IAV surveillance in wild birds.
Collapse
Affiliation(s)
- Yinfeng Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lu Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Shantou University Medical College, Shantou 515041, China
| | - Minsha Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Runyu Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Key Laboratory for Repository and Application of Pathogenic Microbiology, Research Center for Pathogens Detection Technology of Emerging Infectious Diseases, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510000, China
| | - Can Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Shantou University Medical College, Shantou 515041, China
| | - Yangtong Tan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Pei Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Dan Xiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Shantou University Medical College, Shantou 515041, China
| | - Xiaqiong Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yanling Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A8, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, M5S 1A8, Canada
| | - Yongyi Shen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| |
Collapse
|
15
|
Tundup S, Kandasamy M, Perez JT, Mena N, Steel J, Nagy T, Albrecht RA, Manicassamy B. Endothelial cell tropism is a determinant of H5N1 pathogenesis in mammalian species. PLoS Pathog 2017; 13:e1006270. [PMID: 28282445 PMCID: PMC5362246 DOI: 10.1371/journal.ppat.1006270] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 03/22/2017] [Accepted: 03/02/2017] [Indexed: 12/19/2022] Open
Abstract
The cellular and molecular mechanisms underpinning the unusually high virulence of highly pathogenic avian influenza H5N1 viruses in mammalian species remains unknown. Here, we investigated if the cell tropism of H5N1 virus is a determinant of enhanced virulence in mammalian species. We engineered H5N1 viruses with restricted cell tropism through the exploitation of cell type-specific microRNA expression by incorporating microRNA target sites into the viral genome. Restriction of H5N1 replication in endothelial cells via miR-126 ameliorated disease symptoms, prevented systemic viral spread and limited mortality, despite showing similar levels of peak viral replication in the lungs as compared to control virus-infected mice. Similarly, restriction of H5N1 replication in endothelial cells resulted in ameliorated disease symptoms and decreased viral spread in ferrets. Our studies demonstrate that H5N1 infection of endothelial cells results in excessive production of cytokines and reduces endothelial barrier integrity in the lungs, which culminates in vascular leakage and viral pneumonia. Importantly, our studies suggest a need for a combinational therapy that targets viral components, suppresses host immune responses, and improves endothelial barrier integrity for the treatment of highly pathogenic H5N1 virus infections. In healthy individuals, the symptoms of seasonal influenza virus infection are mild and the infection is cleared within 4–7 days. However, infection with highly pathogenic avian influenza virus (H5N1) can be severe and often results in fatal pneumonia even in healthy adults. While it is known that both viral and host factors play a role in enhanced disease progression, the molecular mechanisms for the high virulence of H5N1 virus are not completely understood. In this study, we engineered avian influenza H5N1 viruses incapable of replicating in endothelial cells and evaluated disease symptoms in mice and ferrets. Our studies show that H5N1 infection of endothelial cells causes severe disease and death of infected animals in part due to the damage of endothelial cells lining the blood vessels, which results in leakage of fluid into the lungs (pneumonia).
Collapse
Affiliation(s)
- Smanla Tundup
- Department of Microbiology, University of Chicago, Chicago, IL, United States of America
- Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, IL, United States of America
| | - Matheswaran Kandasamy
- Department of Microbiology, University of Chicago, Chicago, IL, United States of America
- Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, IL, United States of America
| | - Jasmine T. Perez
- Department of Microbiology, University of Chicago, Chicago, IL, United States of America
| | - Nacho Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - John Steel
- Department of Microbiology, Emory University, Atlanta, GA, United States of America
| | - Tamas Nagy
- Comparative Pathology Laboratory, University of Georgia, Athens, GA, United States of America
| | - Randy A. Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Balaji Manicassamy
- Department of Microbiology, University of Chicago, Chicago, IL, United States of America
- Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, IL, United States of America
- * E-mail:
| |
Collapse
|
16
|
Immunogenicity and Cross Protection in Mice Afforded by Pandemic H1N1 Live Attenuated Influenza Vaccine Containing Wild-Type Nucleoprotein. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9359276. [PMID: 28210631 PMCID: PMC5292185 DOI: 10.1155/2017/9359276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/21/2016] [Indexed: 01/31/2023]
Abstract
Since conserved viral proteins of influenza virus, such as nucleoprotein (NP) and matrix 1 protein, are the main targets for virus-specific CD8+ cytotoxic T-lymphocytes (CTLs), we hypothesized that introduction of the NP gene of wild-type virus into the genome of vaccine reassortants could lead to better immunogenicity and afford better protection. This paper describes in vitro and in vivo preclinical studies of two new reassortants of pandemic H1N1 live attenuated influenza vaccine (LAIV) candidates. One had the hemagglutinin (HA) and neuraminidase (NA) genes from A/South Africa/3626/2013 H1N1 wild-type virus on the A/Leningrad/134/17/57 master donor virus backbone (6 : 2 formulation) while the second had the HA, NA, and NP genes of the wild-type virus on the same backbone (5 : 3 formulation). Although both LAIVs induced similar antibody immune responses, the 5 : 3 LAIV provoked greater production of virus-specific CTLs than the 6 : 2 variant. Furthermore, the 5 : 3 LAIV-induced CTLs had higher in vivo cytotoxic activity, compared to 6 : 2 LAIV. Finally, the 5 : 3 LAIV candidate afforded greater protection against infection and severe illness than the 6 : 2 LAIV. Inclusion in LAIV of the NP gene from wild-type influenza virus is a new approach to inducing cross-reactive cell-mediated immune responses and cross protection against pandemic influenza.
Collapse
|
17
|
Perwitasari O, Johnson S, Yan X, Register E, Crabtree J, Gabbard J, Howerth E, Shacham S, Carlson R, Tamir S, Tripp RA. Antiviral Efficacy of Verdinexor In Vivo in Two Animal Models of Influenza A Virus Infection. PLoS One 2016; 11:e0167221. [PMID: 27893810 PMCID: PMC5125695 DOI: 10.1371/journal.pone.0167221] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/10/2016] [Indexed: 11/26/2022] Open
Abstract
Influenza A virus (IAV) causes seasonal epidemics of respiratory illness that can cause mild to severe illness and potentially death. Antiviral drugs are an important countermeasure against IAV; however, drug resistance has developed, thus new therapeutic approaches are being sought. Previously, we demonstrated the antiviral activity of a novel nuclear export inhibitor drug, verdinexor, to reduce influenza replication in vitro and pulmonary virus burden in mice. In this study, in vivo efficacy of verdinexor was further evaluated in two animal models or influenza virus infection, mice and ferrets. In mice, verdinexor was efficacious to limit virus shedding, reduce pulmonary pro-inflammatory cytokine expression, and moderate leukocyte infiltration into the bronchoalveolar space. Similarly, verdinexor-treated ferrets had reduced lung pathology, virus burden, and inflammatory cytokine expression in the nasal wash exudate. These findings support the anti-viral efficacy of verdinexor, and warrant its development as a novel antiviral therapeutic for influenza infection.
Collapse
Affiliation(s)
- Olivia Perwitasari
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Scott Johnson
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Xiuzhen Yan
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Emery Register
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Jackelyn Crabtree
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Jon Gabbard
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Elizabeth Howerth
- Department of Pathology, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| | - Sharon Shacham
- Karyopharm Therapeutics, Newton, Massachusetts, United States of America
| | - Robert Carlson
- Karyopharm Therapeutics, Newton, Massachusetts, United States of America
| | - Sharon Tamir
- Karyopharm Therapeutics, Newton, Massachusetts, United States of America
| | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, United States of America
| |
Collapse
|
18
|
Pathogenesis and Transmission Assessments of Two H7N8 Influenza A Viruses Recently Isolated from Turkey Farms in Indiana Using Mouse and Ferret Models. J Virol 2016; 90:10936-10944. [PMID: 27681133 DOI: 10.1128/jvi.01646-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/21/2016] [Indexed: 01/14/2023] Open
Abstract
Avian influenza A H7 viruses have caused multiple outbreaks in domestic poultry throughout North America, resulting in occasional infections of humans in close contact with affected birds. In early 2016, the presence of H7N8 highly pathogenic avian influenza (HPAI) viruses and closely related H7N8 low-pathogenic avian influenza (LPAI) viruses was confirmed in commercial turkey farms in Indiana. These H7N8 viruses represent the first isolation of this subtype in domestic poultry in North America, and their virulence in mammalian hosts and the potential risk for human infection are largely unknown. In this study, we assessed the ability of H7N8 HPAI and LPAI viruses to replicate in vitro in human airway cells and in vivo in mouse and ferret models. Both H7N8 viruses replicated efficiently in vitro and in vivo, but they exhibited substantial differences in disease severity in mammals. In mice, while the H7N8 LPAI virus largely remained avirulent, the H7N8 HPAI virus exhibited greater infectivity, virulence, and lethality. Both H7N8 viruses replicated similarly in ferrets, but only the H7N8 HPAI virus caused moderate weight loss, lethargy, and mortality. The H7N8 LPAI virus displayed limited transmissibility in ferrets placed in direct contact with an inoculated animal, while no transmission of H7N8 HPAI virus was detected. Our results indicate that the H7N8 avian influenza viruses from Indiana are able to replicate in mammals and cause severe disease but with limited transmission. The recent appearance of H7N8 viruses in domestic poultry highlights the need for continued influenza surveillance in wild birds and close monitoring of the potential risk to human health. IMPORTANCE H7 influenza viruses circulate in wild birds in the United States, but when the virus emerges in domestic poultry populations, the frequency of human exposure and the potential for human infections increases. An H7N8 highly pathogenic avian influenza (HPAI) virus and an H7N8 low-pathogenic avian influenza (LPAI) virus were recently isolated from commercial turkey farms in Indiana. To determine the risk that these influenza viruses pose to humans, we assessed their pathogenesis and transmission in vitro and in mammalian models. We found that the H7N8 HPAI virus exhibited enhanced virulence, and although transmission was only observed with the H7N8 LPAI virus, the ability of this H7 virus to transmit in a mammalian host and quickly evolve to a more virulent strain is cause for concern. Our findings offer important insight into the potential for emerging H7 avian influenza viruses to acquire the ability to cause disease and transmit among mammals.
Collapse
|
19
|
Lipsitch M, Barclay W, Raman R, Russell CJ, Belser JA, Cobey S, Kasson PM, Lloyd-Smith JO, Maurer-Stroh S, Riley S, Beauchemin CA, Bedford T, Friedrich TC, Handel A, Herfst S, Murcia PR, Roche B, Wilke CO, Russell CA. Viral factors in influenza pandemic risk assessment. eLife 2016; 5. [PMID: 27834632 PMCID: PMC5156527 DOI: 10.7554/elife.18491] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022] Open
Abstract
The threat of an influenza A virus pandemic stems from continual virus spillovers from reservoir species, a tiny fraction of which spark sustained transmission in humans. To date, no pandemic emergence of a new influenza strain has been preceded by detection of a closely related precursor in an animal or human. Nonetheless, influenza surveillance efforts are expanding, prompting a need for tools to assess the pandemic risk posed by a detected virus. The goal would be to use genetic sequence and/or biological assays of viral traits to identify those non-human influenza viruses with the greatest risk of evolving into pandemic threats, and/or to understand drivers of such evolution, to prioritize pandemic prevention or response measures. We describe such efforts, identify progress and ongoing challenges, and discuss three specific traits of influenza viruses (hemagglutinin receptor binding specificity, hemagglutinin pH of activation, and polymerase complex efficiency) that contribute to pandemic risk.
Collapse
Affiliation(s)
- Marc Lipsitch
- Center for Communicable Disease Dynamics, Harvard T. H Chan School of Public Health, Boston, United States.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States
| | - Wendy Barclay
- Division of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Rahul Raman
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Charles J Russell
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, United States
| | - Jessica A Belser
- Centers for Disease Control and Prevention, Atlanta, United States
| | - Sarah Cobey
- Department of Ecology and Evolutionary Biology, University of Chicago, Chicago, United States
| | - Peter M Kasson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, United States.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - James O Lloyd-Smith
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, United States.,Fogarty International Center, National Institutes of Health, Bethesda, United States
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, Agency for Science Technology and Research, Singapore, Singapore.,National Public Health Laboratory, Communicable Diseases Division, Ministry of Health, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Steven Riley
- MRC Centre for Outbreak Analysis and Modelling, School of Public Health, Imperial College London, London, United Kingdom.,Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | | | - Trevor Bedford
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, United States
| | - Andreas Handel
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, United States
| | - Sander Herfst
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Pablo R Murcia
- MRC-University of Glasgow Centre For Virus Research, Glasgow, United Kingdom
| | | | - Claus O Wilke
- Center for Computational Biology and Bioinformatics, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, United States.,Department of Integrative Biology, The University of Texas at Austin, Austin, United States
| | - Colin A Russell
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
20
|
Kim SM, Kim YI, Pascua PNQ, Choi YK. Avian Influenza A Viruses: Evolution and Zoonotic Infection. Semin Respir Crit Care Med 2016; 37:501-11. [PMID: 27486732 PMCID: PMC7171714 DOI: 10.1055/s-0036-1584953] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although efficient human-to-human transmission of avian influenza virus has yet to be seen, in the past two decades avian-to-human transmission of influenza A viruses has been reported. Influenza A/H5N1, in particular, has repeatedly caused human infections associated with high mortality, and since 1998 the virus has evolved into many clades of variants with significant antigenic diversity. In 2013, three (A/H7N9, A/H6N1, and A/H10N8) novel avian influenza viruses (AIVs) breached the animal-human host species barrier in Asia. In humans, roughly 35% of A/H7N9-infected patients succumbed to the zoonotic infection, and two of three A/H10N8 human infections were also lethal; however, neither of these viruses cause influenza-like symptoms in poultry. While most of these cases were associated with direct contact with infected poultry, some involved sustained human-to-human transmission. Thus, these events elicited concern regarding potential AIV pandemics. This article reviews the human incursions associated with AIV variants and the potential role of pigs as an intermediate host that may hasten AIV evolution. In addition, we discuss the known influenza A virus virulence and transmission factors and their evaluation in animal models. With the growing number of human AIV infections, constant vigilance for the emergence of novel viruses is of utmost importance. In addition, careful characterization and pathobiological assessment of these novel variants will help to identify strains of particular concern for future pandemics.
Collapse
Affiliation(s)
- Se Mi Kim
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Young-Il Kim
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Philippe Noriel Q Pascua
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Young Ki Choi
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
21
|
Jiao P, Cui J, Song Y, Song H, Zhao Z, Wu S, Qu N, Wang N, Ouyang G, Liao M. New Reassortant H5N6 Highly Pathogenic Avian Influenza Viruses in Southern China, 2014. Front Microbiol 2016; 7:754. [PMID: 27242767 PMCID: PMC4872003 DOI: 10.3389/fmicb.2016.00754] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/04/2016] [Indexed: 01/23/2023] Open
Abstract
New reassortant H5N6 highly pathogenic avian influenza viruses (AIVs) were isolated from apparently healthy domestic ducks in Southern China in 2014. Our results show that the viruses grew efficiently in eggs and replicated systemically in chickens. They were completely lethal in chicken (100% mortality), and the mean death time was 6 to 7 days post-inoculation. The viruses could transmit in chickens by naïve contact. BLAST analysis revealed that their HA gene was most closely related to A/wild duck/Shangdong/628/2011 (H5N1), and their NA genes were most closely related to A/swine/Guangdong/K6/2010 (H6N6). The other genes had the highest identity with A/wild duck/Fujian/1/2011(H5N1). The results of phylogenetic analysis showed that their HA genes clustered into clade 2.3.4.4 of the H5N1 viruses and all genes derived from H5 were Mix-like or H6-like viruses. Thus, the new H5N6 viruses were reassortmented of H5N1 and H6N6 virus. Therefore, the circulation of the new H5N6 AIVs may become a threat to poultry and human health.
Collapse
Affiliation(s)
- Peirong Jiao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Jin Cui
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Yafen Song
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Hui Song
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Zhishan Zhao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; College of Life Science, South China Agricultural UniversityGuangzhou, China
| | - Siyu Wu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Nannan Qu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Nianchen Wang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Guowen Ouyang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and ControlGuangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of AgricultureGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of GuangdongGuangzhou, China; College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| |
Collapse
|
22
|
Job ER, Pizzolla A, Nebl T, Short KR, Deng YM, Carolan L, Laurie KL, Brooks AG, Reading PC. Neutralizing inhibitors in the airways of naïve ferrets do not play a major role in modulating the virulence of H3 subtype influenza A viruses. Virology 2016; 494:143-57. [PMID: 27110707 DOI: 10.1016/j.virol.2016.01.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 12/09/2022]
Abstract
Many insights regarding the pathogenesis of human influenza A virus (IAV) infections have come from studies in mice and ferrets. Surfactant protein (SP)-D is the major neutralizing inhibitor of IAV in mouse airway fluids and SP-D-resistant IAV mutants show enhanced virus replication and virulence in mice. Herein, we demonstrate that sialylated glycoproteins, rather than SP-D, represent the major neutralizing inhibitors against H3 subtype viruses in airway fluids from naïve ferrets. Moreover, while resistance to neutralizing inhibitors is a critical factor in modulating virus replication and disease in the mouse model, it does not appear to be so in the ferret model, as H3 mutants resistant to either SP-D or sialylated glycoproteins in ferret airway fluids did not show enhanced virulence in ferrets. These data have important implications for our understanding of pathogenesis and immunity to human IAV infections in these two widely used animal models of infection.
Collapse
Affiliation(s)
- Emma R Job
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Angela Pizzolla
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Thomas Nebl
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Kirsty R Short
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Yi-Mo Deng
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Karen L Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
23
|
Cox F, Baart M, Huizingh J, Tolboom J, Dekking L, Goudsmit J, Saeland E, Radošević K. Protection against H5N1 Influenza Virus Induced by Matrix-M Adjuvanted Seasonal Virosomal Vaccine in Mice Requires Both Antibodies and T Cells. PLoS One 2015; 10:e0145243. [PMID: 26696245 PMCID: PMC4687931 DOI: 10.1371/journal.pone.0145243] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/30/2015] [Indexed: 12/04/2022] Open
Abstract
Background It remains important to develop the next generation of influenza vaccines that can provide protection against vaccine mismatched strains and to be prepared for potential pandemic outbreaks. To achieve this, the understanding of the immunological parameters that mediate such broad protection is crucial. Method In the current study we assessed the contribution of humoral and cellular immune responses to heterosubtypic protection against H5N1 induced by a Matrix-M (MM) adjuvanted seasonal influenza vaccine by serum transfer and T-cell depletion studies. Results We demonstrate that the heterosubtypic protection against H5N1 induced by MM adjuvanted vaccine is partially mediated by antibodies. The serum contained both H5N1 cross-reactive hemagglutinin (HA)- and neuraminidase (NA)-specific antibodies but with limited virus neutralizing and no hemagglutination inhibiting activity. The cross-reactive antibodies induced antibody-dependent cellular cytotoxicity (ADCC) in vitro, suggesting a role for the Fc part of the antibodies in protection against H5N1. Besides H5N1 specific antibody responses, cross-reactive HA- and NA-specific T-cell responses were induced by the adjuvanted vaccine. T-cell depletion experiments demonstrated that both CD4+ and CD8+ T cells contribute to protection. Conclusion Our study demonstrates that cross-protection against H5N1 induced by MM adjuvanted seasonal virosomal influenza vaccine requires both the humoral and cellular arm of the immune system.
Collapse
Affiliation(s)
- Freek Cox
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Matthijs Baart
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Jeroen Huizingh
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Jeroen Tolboom
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Liesbeth Dekking
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Jaap Goudsmit
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Eirikur Saeland
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
- * E-mail:
| | - Katarina Radošević
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| |
Collapse
|
24
|
Pavulraj S, Bera BC, Joshi A, Anand T, Virmani M, Vaid RK, Shanmugasundaram K, Gulati BR, Rajukumar K, Singh R, Misri J, Singh RK, Tripathi BN, Virmani N. Pathology of Equine Influenza virus (H3N8) in Murine Model. PLoS One 2015; 10:e0143094. [PMID: 26587990 PMCID: PMC4654517 DOI: 10.1371/journal.pone.0143094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/30/2015] [Indexed: 01/09/2023] Open
Abstract
Equine influenza viruses (EIV)—H3N8 continue to circulate in equine population throughout the world. They evolve by the process of antigenic drift that leads to substantial change in the antigenicity of the virus, thereby necessitating substitution of virus strain in the vaccines. This requires frequent testing of the new vaccines in the in vivo system; however, lack of an appropriate laboratory animal challenge model for testing protective efficacy of equine influenza vaccine candidates hinders the screening of new vaccines and other therapeutic approaches. In the present investigation, BALB/c mouse were explored for suitability for conducting pathogenecity studies for EIV. The BALB/c mice were inoculated intranasally @ 2×106.24 EID50 with EIV (H3N8) belonging to Clade 2 of Florida sublineage and monitored for setting up of infection and associated parameters. All mice inoculated with EIV exhibited clinical signs viz. loss in body weights, lethargy, dyspnea, etc, between 3 and 5 days which commensurate with lesions observed in the respiratory tract including rhinitis, tracheitis, bronchitis, bronchiolitis, alveolitis and diffuse interstitial pneumonia. Transmission electron microscopy, immunohistochemistry, virus quantification through titration and qRT-PCR demonstrated active viral infection in the upper and lower respiratory tract. Serology revealed rise in serum lactate dehydrogenase levels along with sero-conversion. The pattern of disease progression, pathological lesions and virus recovery from nasal washings and lungs in the present investigations in mice were comparable to natural and experimental EIV infection in equines. The findings establish BALB/c mice as small animal model for studying EIV (H3N8) infection and will have immense potential for dissecting viral pathogenesis, vaccine efficacy studies, preliminary screening of vaccine candidates and antiviral therapeutics against EIV.
Collapse
Affiliation(s)
| | | | - Alok Joshi
- Veterinary Hospital—Naini, Barakot, Almora, Uttarakhand, India
| | - Taruna Anand
- National Research Centre on Equines, Hisar, Haryana, India
| | - Meenakshi Virmani
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary & Animal Sciences, Hisar, Haryana, India
| | | | | | | | - K. Rajukumar
- National Institute of High Security Animal Diseases, Bhopal, MP, India
| | - Rajendra Singh
- Division of Pathology, Indian Veterinary Research Institute, Bareilly, UP, India
| | - Jyoti Misri
- Division of Animal Science, Krishi Bhavan, New Delhi, India
| | | | | | - Nitin Virmani
- National Research Centre on Equines, Hisar, Haryana, India
- * E-mail:
| |
Collapse
|
25
|
Cox F, Roos A, Hafkemeijer N, Baart M, Tolboom J, Dekking L, Stittelaar K, Goudsmit J, Radošević K, Saeland E. Matrix-M Adjuvated Seasonal Virosomal Influenza Vaccine Induces Partial Protection in Mice and Ferrets against Avian H5 and H7 Challenge. PLoS One 2015; 10:e0135723. [PMID: 26402787 PMCID: PMC4581625 DOI: 10.1371/journal.pone.0135723] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/24/2015] [Indexed: 11/19/2022] Open
Abstract
There is a constant threat of zoonotic influenza viruses causing a pandemic outbreak in humans. It is virtually impossible to predict which virus strain will cause the next pandemic and it takes a considerable amount of time before a safe and effective vaccine will be available once a pandemic occurs. In addition, development of pandemic vaccines is hampered by the generally poor immunogenicity of avian influenza viruses in humans. An effective pre-pandemic vaccine is therefore required as a first line of defense. Broadening of the protective efficacy of current seasonal vaccines by adding an adjuvant may be a way to provide such first line of defense. Here we evaluate whether a seasonal trivalent virosomal vaccine (TVV) adjuvated with the saponin-based adjuvant Matrix-M (MM) can confer protection against avian influenza H5 and H7 virus strains in mice and ferrets. We demonstrate that mice were protected from death against challenges with H5N1 and H7N7, but that the protection was not complete as evidenced by severe clinical signs. In ferrets, protection against H7N9 was not observed. In contrast, reduced upper and lower respiratory tract viral loads and reduced lung pathology, was achieved in H5N1 challenged ferrets. Together these results suggest that, at least to some extent, Matrix-M adjuvated seasonal virosomal influenza vaccine can serve as an interim measure to decrease morbidity and mortality associated with a pandemic outbreak.
Collapse
Affiliation(s)
- Freek Cox
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Anna Roos
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Nicole Hafkemeijer
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Matthijs Baart
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Jeroen Tolboom
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Liesbeth Dekking
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | | | - Jaap Goudsmit
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Katarina Radošević
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
| | - Eirikur Saeland
- Janssen Prevention Center, Center of Excellence of Janssen Research & Development, Pharmaceutical companies of Johnson and Johnson, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
26
|
Thor SW, Nguyen H, Balish A, Hoang AN, Gustin KM, Nhung PT, Jones J, Thu NN, Davis W, Ngoc TNT, Jang Y, Sleeman K, Villanueva J, Kile J, Gubareva LV, Lindstrom S, Tumpey TM, Davis CT, Long NT. Detection and Characterization of Clade 1 Reassortant H5N1 Viruses Isolated from Human Cases in Vietnam during 2013. PLoS One 2015; 10:e0133867. [PMID: 26244768 PMCID: PMC4526568 DOI: 10.1371/journal.pone.0133867] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/03/2015] [Indexed: 02/03/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 is endemic in Vietnamese poultry and has caused sporadic human infection in Vietnam since 2003. Human infections with HPAI H5N1 are of concern due to a high mortality rate and the potential for the emergence of pandemic viruses with sustained human-to-human transmission. Viruses isolated from humans in southern Vietnam have been classified as clade 1 with a single genome constellation (VN3) since their earliest detection in 2003. This is consistent with detection of this clade/genotype in poultry viruses endemic to the Mekong River Delta and surrounding regions. Comparison of H5N1 viruses detected in humans from southern Vietnamese provinces during 2012 and 2013 revealed the emergence of a 2013 reassortant virus with clade 1.1.2 hemagglutinin (HA) and neuraminidase (NA) surface protein genes but internal genes derived from clade 2.3.2.1a viruses (A/Hubei/1/2010-like; VN12). Closer analysis revealed mutations in multiple genes of this novel genotype (referred to as VN49) previously associated with increased virulence in animal models and other markers of adaptation to mammalian hosts. Despite the changes identified between the 2012 and 2013 genotypes analyzed, their virulence in a ferret model was similar. Antigenically, the 2013 viruses were less cross-reactive with ferret antiserum produced to the clade 1 progenitor virus, A/Vietnam/1203/2004, but reacted with antiserum produced against a new clade 1.1.2 WHO candidate vaccine virus (A/Cambodia/W0526301/2012) with comparable hemagglutination inhibition titers as the homologous antigen. Together, these results indicate changes to both surface and internal protein genes of H5N1 viruses circulating in southern Vietnam compared to 2012 and earlier viruses.
Collapse
Affiliation(s)
- Sharmi W. Thor
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Hieu Nguyen
- Institute Pasteur-Ho Chi Minh City, National Influenza Center-2, Ho Chi Minh City, Vietnam
| | - Amanda Balish
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Anh Nguyen Hoang
- Institute Pasteur-Ho Chi Minh City, National Influenza Center-2, Ho Chi Minh City, Vietnam
| | - Kortney M. Gustin
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Pham Thi Nhung
- Institute Pasteur-Ho Chi Minh City, National Influenza Center-2, Ho Chi Minh City, Vietnam
| | - Joyce Jones
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ngoc Nguyen Thu
- Institute Pasteur-Ho Chi Minh City, National Influenza Center-2, Ho Chi Minh City, Vietnam
| | - William Davis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Thao Nguyen Thi Ngoc
- Institute Pasteur-Ho Chi Minh City, National Influenza Center-2, Ho Chi Minh City, Vietnam
| | - Yunho Jang
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Katrina Sleeman
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Julie Villanueva
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James Kile
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Influenza Program, Centers for Disease Control and Prevention- Vietnam, Hanoi, Vietnam
| | - Larisa V. Gubareva
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Stephen Lindstrom
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Terrence M. Tumpey
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - C. Todd Davis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (NTL); (CTD)
| | - Nguyen Thanh Long
- Institute Pasteur-Ho Chi Minh City, National Influenza Center-2, Ho Chi Minh City, Vietnam
- * E-mail: (NTL); (CTD)
| |
Collapse
|
27
|
Recombinant H7 hemagglutinin forms subviral particles that protect mice and ferrets from challenge with H7N9 influenza virus. Vaccine 2015. [PMID: 26207590 DOI: 10.1016/j.vaccine.2015.07.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel avian-origin influenza A H7N9 virus emerged in China in 2013 and continues to cause sporadic human infections with mortality rates approaching 35%. Currently there are no approved human vaccines for H7N9 virus. Recombinant approaches including hemagglutinin (HA) and virus-like particles (VLPs) have resulted in experimental vaccines with advantageous safety and manufacturing characteristics. While high immunogenicity of VLP vaccines has been attributed to the native conformation of HA arranged in the regular repeated patterns within virus-like structures, there is limited data regarding molecular organization of HA within recombinant HA vaccine preparations. In this study, the full-length recombinant H7 protein (rH7) of A/Anhui/1/2013 (H7N9) virus was expressed in Sf9 cells. We showed that purified full-length rH7 retained functional ability to agglutinate red blood cells and formed oligomeric pleomorphic subviral particles (SVPs) of ∼20nm in diameter composed of approximately 10 HA0 molecules. No significant quantities of free monomeric HA0 were observed in rH7 preparation by size exclusion chromatography. Immunogenicity and protective efficacy of rH7 SVPs was confirmed in the mouse and ferret challenge models suggesting that SVPs can be used for vaccination against H7N9 virus.
Collapse
|
28
|
Animal models for influenza virus transmission studies: a historical perspective. Curr Opin Virol 2015; 13:101-8. [PMID: 26126082 DOI: 10.1016/j.coviro.2015.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/10/2015] [Indexed: 01/09/2023]
Abstract
Animal models are used to simulate, under experimental conditions, the complex interactions among host, virus, and environment that affect the person-to-person spread of influenza viruses. The three species that have been most frequently employed, both past and present, as influenza virus transmission models-ferrets, mice, and guinea pigs-have each provided unique insights into the factors governing the efficiency with which these viruses pass from an infected host to a susceptible one. This review will highlight a few of these noteworthy discoveries, with a particular focus on the historical contexts in which each model was developed and the advantages and disadvantages of each species with regard to the study of influenza virus transmission among mammals.
Collapse
|
29
|
Preusse M, Schughart K, Wilk E, Klawonn F, Pessler F. Hematological parameters in the early phase of influenza A virus infection in differentially susceptible inbred mouse strains. BMC Res Notes 2015; 8:225. [PMID: 26047817 PMCID: PMC4467623 DOI: 10.1186/s13104-015-1195-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/20/2015] [Indexed: 12/29/2022] Open
Abstract
Background Hematological parameters have not received much attention in small animal models of infection, particularly at very early time points. We therefore studied changes in leukocyte and thrombocyte numbers in a mouse model of influenza A virus (IAV) infection, including measurements within the first 24 h after infection, and also assessing effects, if any, of the infection/anesthesia procedure on these parameters. Methods DBA/2J and C57BL/6J mice (n = 5–8 per observation) were evaluated in a time course experiment of IAV infection, focusing on early time points. After anesthesia with ketamine/xylazine, a suspension of 2 × 103 focus forming units of the mouse-adapted IAV strain A/Puerto Rico/8/1934 (H1N1) in 20 µl sterile PBS, or 20 µl sterile PBS only (“mock treatment”), were instilled intranasally. Weight loss was assessed daily, and eight common hematological parameters and viral hemagglutinin (HA) mRNA expression were determined after 6, 12, 18, 24, 48 and 120 h. Results Hematological differences between the strains were apparent even in untreated mice. Infection-dependent changes, in particular increased granulocyte and decreased lymphocyte counts, were first detectable after 18 h in DBA/2J, were fully manifest in both strains at 48 h, and were usually more pronounced in the DBA/2J mice. In this strain, relative granulocyte and lymphocyte counts and the granulocyte/lymphocyte ratio correlated with viral HA mRNA expression and weight loss. In C57BL/6J, hematological parameters did not correlate with weight loss, but HA mRNA expression correlated weakly with total leukocyte counts, granulocyte/lymphocyte ratio, relative and absolute granulocyte counts, and relative lymphocyte counts. Significant changes due to mock treatment were mild and were detected only in C57BL/6J. Conclusion This study underscores the value of hematological parameters in monitoring disease evolution in the early phase of IAV infection, and likely other pathogens. The hematological response to infection may differ significantly among inbred mouse strains.
Collapse
Affiliation(s)
- Matthias Preusse
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany. .,Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany.
| | - Klaus Schughart
- Bioinformatics, Helmholtz Centre for Infection Research, Brunswick, Germany. .,University of Veterinary Medicine, Hannover, Germany. .,University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Esther Wilk
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Brunswick, Germany.
| | - Frank Klawonn
- Bioinformatics, Helmholtz Centre for Infection Research, Brunswick, Germany.
| | - Frank Pessler
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany. .,Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany.
| |
Collapse
|
30
|
Repeated Low-Dose Influenza Virus Infection Causes Severe Disease in Mice: a Model for Vaccine Evaluation. J Virol 2015; 89:7841-51. [PMID: 25995265 DOI: 10.1128/jvi.00976-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/11/2015] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Influenza infection causes severe disease and death in humans. In traditional vaccine research and development, a single high-dose virus challenge of animals is used to evaluate vaccine efficacy. This type of challenge model may have limitations. In the present study, we developed a novel challenge model by infecting mice repeatedly in short intervals with low doses of influenza A virus. Our results show that compared to a single high-dose infection, mice that received repeated low-dose challenges showed earlier morbidity and mortality and more severe disease. They developed higher vial loads, more severe lung pathology, and greater inflammatory responses and generated only limited influenza A virus-specific B and T cell responses. A commercial trivalent influenza vaccine protected mice against a single high and lethal dose of influenza A virus but was ineffective against repeated low-dose virus challenges. Overall, our data show that the repeated low-dose influenza A virus infection mouse model is more stringent and may thus be more suitable to select for highly efficacious influenza vaccines. IMPORTANCE Influenza epidemics and pandemics pose serious threats to public health. Animal models are crucial for evaluating the efficacy of influenza vaccines. Traditional models based on a single high-dose virus challenge may have limitations. Here, we describe a new mouse model based on repeated low-dose influenza A virus challenges given within a short period. Repeated low-dose challenges caused more severe disease in mice, associated with higher viral loads and increased lung inflammation and reduced influenza A virus-specific B and T cell responses. A commercial influenza vaccine that was shown to protect mice from high-dose challenge was ineffective against repeated low-dose challenges. Overall, our results show that the low-dose repeated-challenge model is more stringent and may therefore be better suited for preclinical vaccine efficacy studies.
Collapse
|
31
|
Enkirch T, von Messling V. Ferret models of viral pathogenesis. Virology 2015; 479-480:259-70. [PMID: 25816764 PMCID: PMC7111696 DOI: 10.1016/j.virol.2015.03.017] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 01/28/2015] [Accepted: 03/02/2015] [Indexed: 11/26/2022]
Abstract
Emerging and well-known viral diseases remain one the most important global public health threats. A better understanding of their pathogenesis and mechanisms of transmission requires animal models that accurately reproduce these aspects of the disease. Here we review the role of ferrets as an animal model for the pathogenesis of different respiratory viruses with an emphasis on influenza and paramyxoviruses. We will describe the anatomic and physiologic characteristics that contribute to the natural susceptibility of ferrets to these viruses, and provide an overview of the approaches available to analyze their immune responses. Recent insights gained using this model will be highlighted, including the development of new prophylactic and therapeutic approaches. To provide decision criteria for the use of this animal model, its strengths and limitations will be discussed. Ferrets as models for respiratory virus pathogenesis. Ferrets as models for vaccine and drug efficacy assessment. Immunological tools for ferrets. Housing and handling of ferrets.
Collapse
Affiliation(s)
- T Enkirch
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - V von Messling
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany.
| |
Collapse
|
32
|
Comparison of traditional intranasal and aerosol inhalation inoculation of mice with influenza A viruses. Virology 2015; 481:107-12. [PMID: 25771498 DOI: 10.1016/j.virol.2015.02.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/08/2015] [Accepted: 02/18/2015] [Indexed: 12/22/2022]
Abstract
Intranasal instillation of virus in a liquid suspension (IN) is the most frequently employed method to inoculate small mammalian models with influenza virus, but does not reflect a natural route of exposure. In contrast, inoculation via aerosol inhalation (AR) more closely resembles human exposure to influenza virus. Studies in mice have yielded conflicting results regarding virulence induced by virus inoculated by these routes, and have not controlled for potential strain-specific differences, or examined contemporary influenza viruses and avian viruses with pandemic potential. We used a whole-body AR inoculation method to compare infectivity and disease progression of a highly pathogenic H5N1, a low pathogenic H7N9, and a 2009 H1N1 virus with traditional IN inoculation in the mouse model. Generally comparable levels of morbidity and mortality were observed with all viruses examined using either inoculation route, indicating that both IN and AR delivery are appropriate for murine studies investigating influenza virus pathogenicity.
Collapse
|
33
|
Liu YV, Massare MJ, Pearce MB, Sun X, Belser JA, Maines TR, Creager HM, Glenn GM, Pushko P, Smith GE, Tumpey TM. Recombinant virus-like particles elicit protective immunity against avian influenza A(H7N9) virus infection in ferrets. Vaccine 2015; 33:2152-8. [PMID: 25772674 DOI: 10.1016/j.vaccine.2015.03.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/29/2015] [Accepted: 03/03/2015] [Indexed: 12/09/2022]
Abstract
In March 2013, diagnosis of the first reported case of human infection with a novel avian-origin influenza A(H7N9) virus occurred in eastern China. Most human cases have resulted in severe respiratory illness and, in some instances, death. Currently there are no licensed vaccines against H7N9 virus, which continues to cause sporadic human infections. Recombinant virus-like particles (VLPs) have been previously shown to be safe and effective vaccines for influenza. In this study, we evaluated the immunogenicity and protective efficacy of a H7N9 VLP vaccine in the ferret challenge model. Purified recombinant H7N9 VLPs morphologically resembled influenza virions and elicited high-titer serum hemagglutination inhibition (HI) and neutralizing antibodies specific for A/Anhui/1/2013 (H7N9) virus. H7N9 VLP-immunized ferrets subsequently challenged with homologous virus displayed reductions in fever, weight loss, and virus shedding compared to these parameters in unimmunized control ferrets. H7N9 VLP was also effective in protecting against lung and tracheal infection. The addition of either ISCOMATRIX or Matrix-M1 adjuvant improved immunogenicity and protection of the VLP vaccine against H7N9 virus. These results provide support for the development of a safe and effective human VLP vaccine with potent adjuvants against avian influenza H7N9 virus with pandemic potential.
Collapse
Affiliation(s)
- Ye V Liu
- Novavax, Inc., 22 Firstfield, Gaithersburg, MD 20878, USA
| | | | - Melissa B Pearce
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Xiangjie Sun
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Hannah M Creager
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Microbiology and Molecular Genetics Graduate Program, Emory University, Atlanta, GA, USA
| | | | - Peter Pushko
- Medigen, Inc., 8420 Gas House Pike, Frederick, MD, USA
| | - Gale E Smith
- Novavax, Inc., 22 Firstfield, Gaithersburg, MD 20878, USA
| | - Terrence M Tumpey
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
34
|
Rekstin A, Desheva Y, Kiseleva I, Ross T, Swayne D, Rudenko L. Live Attenuated Influenza H7N3 Vaccine is Safe, Immunogenic and Confers Protection in Animal Models. Open Microbiol J 2014; 8:154-62. [PMID: 25685247 PMCID: PMC4323838 DOI: 10.2174/1874285801408010154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/29/2014] [Accepted: 10/02/2014] [Indexed: 02/06/2023] Open
Abstract
Background: In 2003 the outbreak of highly pathogenic H7 avian influenza occurred in the Netherlands. The avian H7 virus causing the outbreak was also detected in humans; one person died of pneumonia and acute respiratory distress syndrome. Our paper describes preclinical studies of a H7N3 live attenuated influenza A vaccine (LAIV) candidate in various animal models. Objectives: To study safety, immunogenicity and protection of H7N3 LAIV candidate in mice, ferrets and chickens. Methods: The vaccine was generated by a classical reassortment between low pathogenicity A/mallard/Netherlands/00 (H7N3) virus and A/Leningrad4/17/57 (H2N2) master donor virus (MDV). Results: Immunogenicity was found that H7N3 LAIV was similar to the MDV in terms of replication in the respiratory organs of mice and failed to replicate in mouse brains. One dose of a H7N3 LAIV elicited measurable antibody response and it was further boosted with a second vaccine dose. Immunization of mice with H7N3 LAIV provided protection against infection following a homologous challenge with wild type H7N3 virus. Attenuated phenotype of H7N3 LAIV has been confirmed in ferrets. Immunogenicity and protective efficacy of H7N3 LAIV in ferrets were also demonstrated. The vaccine protected animals from subsequent infection with wild type H7N3 virus. The results of histopathology study revealed that inoculation of H7N3 LAIV in ferrets did not cause any inflammation or destructive changes in lungs. Lack of H7N3 LAIV replication in chicken demonstrated complete safety of this preparation for poultry. Conclusion: Results of our study suggest that new H7N3 LAIV candidate is safe, immunogenic and protects from homologues influenza virus infection in mice and ferrets.
Collapse
Affiliation(s)
- Andrey Rekstin
- Department of Virology, Institute of Experimental Medicine RAMS, St. Petersburg, Russia
| | - Yulia Desheva
- Department of Virology, Institute of Experimental Medicine RAMS, St. Petersburg, Russia
| | - Irina Kiseleva
- Department of Virology, Institute of Experimental Medicine RAMS, St. Petersburg, Russia
| | - Ted Ross
- University of Pittsburg (Pittsburg, USA)
| | - David Swayne
- Southeast Poultry Research Laboratory (Athens, USA)
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine RAMS, St. Petersburg, Russia
| |
Collapse
|
35
|
Williams D, Basavarajappa MS, Rasmussen SA, Morris S, Mattison D. Highlights from the United States Food and Drug Administration's public workshop on the development of animal models of pregnancy to address medical countermeasures in an "at-risk" population of pregnant women: Influenza as a case study. ACTA ACUST UNITED AC 2014; 100:806-10. [PMID: 25296888 DOI: 10.1002/bdra.23319] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/19/2014] [Accepted: 08/29/2014] [Indexed: 11/08/2022]
Abstract
The U.S. Food and Drug Administration (FDA) and other federal agencies partner to ensure that medical countermeasures (e.g., drug therapies and vaccines) are available for public health emergencies (FDA, 2014). Despite continuing progress, providing medical countermeasures and treatment guidelines for certain populations (e.g., pregnant women) is challenging due to the lack of clinical and/or animal data. Thus, a workshop was convened to discuss animal models of pregnancy for the evaluation of disease progression and medical countermeasures.
Collapse
Affiliation(s)
- Denita Williams
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas; Huntingdon Life Sciences, Inc., Somerset, New Jersey
| | | | | | | | | | | |
Collapse
|
36
|
Adjuvanted inactivated influenza A(H3N2) vaccines induce stronger immunogenicity in mice and confer higher protection in ferrets than unadjuvanted inactivated vaccines. Vaccine 2014; 32:5730-9. [PMID: 25173481 DOI: 10.1016/j.vaccine.2014.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/07/2014] [Accepted: 08/15/2014] [Indexed: 11/20/2022]
Abstract
Influenza viruses are major respiratory pathogens and the development of improved vaccines to prevent these infections is of high priority. Here, we evaluated split inactivated A(H3N2) vaccines (A/Uruguay/716/2007) combined or not with adjuvants (AS03, AS25 and Protollin) and administered by three different routes, intramuscular (i.m.), intranasal (i.n.) or intradermal (i.d.), both in BALB/c mice and in ferrets. Ferrets were challenged with the homologous strain A/Uruguay/716/2007 (H3N2) or the heterologous strain A/Perth/16/2009 (H3N2) 4 weeks after the second immunization with A/Uruguay/716/2007 vaccines. Temperature, weight loss and clinical signs were monitored on a daily basis and nasal washes were performed to evaluate viral titers in the upper respiratory tract. All adjuvanted vaccines induced stronger humoral immune responses than unadjuvanted ones in both mice and ferrets. In mice, the AS03- and AS25-adjuvanted i.m. vaccines generated a mixed Th1-Th2 response at 6 and 19 weeks after the last immunization as shown by the production of IgG1 and IgG2a antibodies as well as the production of IL-2, IL-4 and IFN-γ by CD4+ and CD8+ T cells. HAI and MN titers were also higher in those groups when compared to the i.n. Protollin-adjuvanted and unadjuvanted groups. The Protollin-adjuvanted i.n. vaccine induced a more Th1 oriented response with a significant production of IgA in bronchoalveolar lavages. In ferrets, the AS03- and AS25-adjuvanted i.m. vaccines also induced higher HAI and MN titers compared to the other groups. These vaccines also significantly decreased viral titers after challenge with both the homologous A/Uruguay/716/2007 (H3N2) and the heterologous A/Perth/16/2009 (H3N2) strains. In conclusion, adjuvanted influenza vaccines elicited stronger humoral response in mice and conferred greater protection in naive ferrets than unadjuvanted ones. Interestingly, the AS25 adjuvant system containing monophosphoryl-lipid-A appears particularly promising for developing more potent inactivated influenza vaccines.
Collapse
|
37
|
Jia N, Barclay WS, Roberts K, Yen HL, Chan RWY, Lam AKY, Air G, Peiris JSM, Dell A, Nicholls JM, Haslam SM. Glycomic characterization of respiratory tract tissues of ferrets: implications for its use in influenza virus infection studies. J Biol Chem 2014; 289:28489-504. [PMID: 25135641 PMCID: PMC4192499 DOI: 10.1074/jbc.m114.588541] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The initial recognition between influenza virus and the host cell is mediated by interactions between the viral surface protein hemagglutinin and sialic acid-terminated glycoconjugates on the host cell surface. The sialic acid residues can be linked to the adjacent monosaccharide by α2–3- or α2–6-type glycosidic bonds. It is this linkage difference that primarily defines the species barrier of the influenza virus infection with α2–3 binding being associated with avian influenza viruses and α2–6 binding being associated with human strains. The ferret has been extensively used as an animal model to study the transmission of influenza. To better understand the validity of this model system, we undertook glycomic characterization of respiratory tissues of ferret, which allows a comparison of potential viral receptors to be made between humans and ferrets. To complement the structural analysis, lectin staining experiments were performed to characterize the regional distributions of glycans along the respiratory tract of ferrets. Finally, the binding between the glycans identified and the hemagglutinins of different strains of influenza viruses was assessed by glycan array experiments. Our data indicated that the respiratory tissues of ferret heterogeneously express both α2–3- and α2–6-linked sialic acids. However, the respiratory tissues of ferret also expressed the Sda epitope (NeuAcα2-3(GalNAcβ1–4)Galβ1–4GlcNAc) and sialylated N,N′-diacetyllactosamine (NeuAcα2–6GalNAcβ1–4GlcNAc), which have not been observed in the human respiratory tract surface epithelium. The presence of the Sda epitope reduces potential binding sites for avian viruses and thus may have implications for the usefulness of the ferret in the study of influenza virus infection.
Collapse
Affiliation(s)
- Nan Jia
- From the Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Wendy S Barclay
- the Faculty of Medicine, Division of Infectious Disease, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom
| | - Kim Roberts
- the Faculty of Medicine, Division of Infectious Disease, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom
| | - Hui-Ling Yen
- the School of Public Health, University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Renee W Y Chan
- the School of Public Health, University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Alfred K Y Lam
- the Department of Pathology, Griffith University, 4111 Queensland, Australia
| | - Gillian Air
- the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901, and
| | - J S Malik Peiris
- the School of Public Health, University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Anne Dell
- From the Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - John M Nicholls
- the Department of Pathology, University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Stuart M Haslam
- From the Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom,
| |
Collapse
|
38
|
Dengler L, Kühn N, Shin DL, Hatesuer B, Schughart K, Wilk E. Cellular changes in blood indicate severe respiratory disease during influenza infections in mice. PLoS One 2014; 9:e103149. [PMID: 25058639 PMCID: PMC4110021 DOI: 10.1371/journal.pone.0103149] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/27/2014] [Indexed: 12/23/2022] Open
Abstract
Influenza A infection is a serious threat to human and animal health. Many of the biological mechanisms of the host-pathogen-interactions are still not well understood and reliable biomarkers indicating the course of the disease are missing. The mouse is a valuable model system enabling us to study the local inflammatory host response and the influence on blood parameters under controlled circumstances. Here, we compared the lung and peripheral changes after PR8 (H1N1) influenza A virus infection in C57BL/6J and DBA/2J mice using virus variants of different pathogenicity resulting in non-lethal and lethal disease. We monitored hematological and immunological parameters revealing that the granulocyte to lymphocyte ratio in the blood represents an early indicator of severe disease progression already two days after influenza A infection in mice. These findings might be relevant to optimize early diagnostic options of severe influenza disease and to monitor successful therapeutic treatment in humans.
Collapse
Affiliation(s)
- Leonie Dengler
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nora Kühn
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
| | - Dai-Lun Shin
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
| | - Bastian Hatesuer
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
- University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| | - Esther Wilk
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
39
|
Thangavel RR, Bouvier NM. Animal models for influenza virus pathogenesis, transmission, and immunology. J Immunol Methods 2014; 410:60-79. [PMID: 24709389 PMCID: PMC4163064 DOI: 10.1016/j.jim.2014.03.023] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 12/24/2022]
Abstract
In humans, infection with an influenza A or B virus manifests typically as an acute and self-limited upper respiratory tract illness characterized by fever, cough, sore throat, and malaise. However, influenza can present along a broad spectrum of disease, ranging from sub-clinical or even asymptomatic infection to a severe primary viral pneumonia requiring advanced medical supportive care. Disease severity depends upon the virulence of the influenza virus strain and the immune competence and previous influenza exposures of the patient. Animal models are used in influenza research not only to elucidate the viral and host factors that affect influenza disease outcomes in and spread among susceptible hosts, but also to evaluate interventions designed to prevent or reduce influenza morbidity and mortality in man. This review will focus on the three animal models currently used most frequently in influenza virus research - mice, ferrets, and guinea pigs - and discuss the advantages and disadvantages of each.
Collapse
Affiliation(s)
- Rajagowthamee R Thangavel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Nicole M Bouvier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
40
|
Belser JA, Tumpey TM. Mammalian models for the study of H7 virus pathogenesis and transmission. Curr Top Microbiol Immunol 2014; 385:275-305. [PMID: 24996862 DOI: 10.1007/82_2014_383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mammalian models, most notably the mouse and ferret, have been instrumental in the assessment of avian influenza virus pathogenicity and transmissibility, and have been used widely to characterize the molecular determinants that confer H5N1 virulence in mammals. However, while H7 influenza viruses have typically been associated with conjunctivitis and/or mild respiratory disease in humans, severe disease and death is also possible, as underscored by the recent emergence of H7N9 viruses in China. Despite the public health need to understand the pandemic potential of this virus subtype, H7 virus pathogenesis and transmission has not been as extensively studied. In this review, we discuss the heterogeneity of H7 subtype viruses isolated from humans, and the characterization of mammalian models to study the virulence of H7 subtype viruses associated with human infection, including viruses of both high and low pathogenicity and following multiple inoculation routes. The use of the ferret transmission model to assess the influence of receptor binding preference among contemporary H7 influenza viruses is described. These models have enabled the study of preventative and therapeutic agents, including vaccines and antivirals, to reduce disease burden, and have permitted a greater appreciation that not all highly pathogenic influenza viruses are created equal.
Collapse
Affiliation(s)
- Jessica A Belser
- Influenza Division, MS G-16, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Atlanta, GA, 30333, USA
| | | |
Collapse
|
41
|
Pathogenicity and transmission of H5N1 avian influenza viruses in different birds. Vet Microbiol 2014; 168:50-9. [DOI: 10.1016/j.vetmic.2013.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/12/2013] [Accepted: 10/17/2013] [Indexed: 11/18/2022]
|
42
|
H1N1, but not H3N2, influenza A virus infection protects ferrets from H5N1 encephalitis. J Virol 2013; 88:3077-91. [PMID: 24371072 DOI: 10.1128/jvi.01840-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Seasonal influenza causes substantial morbidity and mortality because of efficient human-to-human spread. Rarely, zoonotic strains of influenza virus spread to humans, where they have the potential to mediate new pandemics with high mortality. We studied systemic viral spread after intranasal infection with highly pathogenic avian influenza virus (H5N1 [A/Viet Nam/1203/2004]) in ferrets with or without prior pandemic H1N1pdm09 (A/Mexico/4108/2009) or H3N2 (A/Victoria/361/2011) infection. After intranasal challenge with H5N1 influenza virus, naive ferrets rapidly succumbed to systemic infection. Animals challenged with H5N1 influenza virus greater than 3 months after recovering from an initial H1N1pdm09 infection survived H5N1 virus challenge and cleared virus from the respiratory tract 4 days after infection. However, a prolonged low-level infection of hematopoietic elements in the small bowel lamina propria, liver, and spleen was present for greater than 2 weeks postinfection, raising the potential for reassortment of influenza genes in a host infected with multiple strains of influenza. Animals previously infected with an H3N2 influenza virus succumbed to systemic disease and encephalitis after H5N1 virus challenge. These results indicate prior infection with different seasonal influenza strains leads to radically different protection from H5N1 challenge and fatal encephalitis. IMPORTANCE Seasonal influenza is efficiently transmitted from human to human, causing substantial morbidity and mortality. Rarely, zoonotic strains of influenza virus spread to humans, where they have the potential to mediate new pandemics with high mortality. Infection of naive ferrets with H5N1 avian influenza virus causes a rapid and lethal systemic disease. We studied systemic H5N1 viral spread after infection of ferrets with or without prior exposure to either of two seasonal influenza virus strains, H1N1 and H3N2. Ferrets previously infected with H1N1 survive H5N1 challenge while those previously infected with H3N2 die of encephalitis. However ferrets protected from lethal H5N1 infection develop persistent low-level infection of the small intestine, liver, or spleen, providing a nidus for future viral strain recombination. The mechanism by which prior infection with specific strains of seasonal influenza virus protect from lethal H5N1 challenge needs to be elucidated in order to design effective immunization and treatments.
Collapse
|
43
|
de Jong RMC, Stockhofe-Zurwieden N, Verheij ES, de Boer-Luijtze EA, Ruiter SJM, de Leeuw OS, Cornelissen LAHM. Rapid emergence of a virulent PB2 E627K variant during adaptation of highly pathogenic avian influenza H7N7 virus to mice. Virol J 2013; 10:276. [PMID: 24007444 PMCID: PMC3766704 DOI: 10.1186/1743-422x-10-276] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 09/02/2013] [Indexed: 11/17/2022] Open
Abstract
Background Highly pathogenic avian influenza (HPAI) viruses pose a potential human health threat as they can be transmitted directly from infected poultry to humans. During a large outbreak of HPAI H7N7 virus among poultry in The Netherlands in 2003, bird to human transmission was confirmed in 89 cases, of which one had a fatal outcome. Methods To identify genetic determinants of virulence in a mammalian host, we passaged an avian H7N7/03 outbreak isolate in mouse lungs and evaluated the phenotype of the mouse-adapted variant in animal models and in vitro. Results Three passages in mouse lungs were sufficient to select a variant that was highly virulent in mice. The virus had a MLD50 that was >4.3 logs lower than that of its non-lethal parental virus. Sequence analysis revealed a single mutation at position 627 in PB2, where the glutamic acid was changed to a lysine (E627K). The mouse-adapted virus has this mutation in common with the fatal human case isolate. The virus remained highly pathogenic for chickens after its passage in mice. In ferrets, the mouse-adapted virus induced more severe disease, replicated to higher titers in the lower respiratory tract and spread more efficiently to systemic organs compared with the parental virus. In vitro, the PB2 E627K mutation had a promoting effect on virus propagation in mammalian, but not in avian cells. Conclusions Our results show that the E627K mutation in PB2 alone can be sufficient to convert an HPAI H7N7 virus of low virulence to a variant causing severe disease in mice and ferrets. The rapid emergence of the PB2 E627K mutant during mouse adaptation and its pathogenicity in ferrets emphasize the potential risk of HPAI H7N7 viruses for human health.
Collapse
|
44
|
Brehm MA, Jouvet N, Greiner DL, Shultz LD. Humanized mice for the study of infectious diseases. Curr Opin Immunol 2013; 25:428-35. [PMID: 23751490 DOI: 10.1016/j.coi.2013.05.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/15/2013] [Accepted: 05/17/2013] [Indexed: 12/17/2022]
Abstract
Many of the pathogens that cause human infectious diseases do not infect rodents or other mammalian species. Small animal models that allow studies of the pathogenesis of these agents and evaluation of drug efficacy are critical for identifying ways to prevent and treat human infectious diseases. Immunodeficient mice engrafted with functional human cells and tissues, termed 'humanized' mice, represent a critical pre-clinical bridge for in vivo studies of human pathogens. Recent advances in the development of humanized mice have allowed in vivo studies of multiple human infectious agents providing novel insights into their pathogenesis that was otherwise not possible.
Collapse
Affiliation(s)
- Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | | | | | | |
Collapse
|
45
|
Rimmelzwaan GF, Katz JM. Immune responses to infection with H5N1 influenza virus. Virus Res 2013; 178:44-52. [PMID: 23735534 DOI: 10.1016/j.virusres.2013.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 04/03/2013] [Accepted: 05/20/2013] [Indexed: 01/22/2023]
Abstract
Influenza A H5N1 viruses remain a substantial threat to global public health. In particular, the expanding genetic diversity of H5N1 viruses and the associated risk for human adaptation underscore the importance of better understanding host immune responses that may protect against disease or infection. Although much emphasis has been placed on investigating early virus-host interactions and the induction of innate immune responses, little is known of the consequent adaptive immune response to H5N1 virus infection. In this review, we describe the H5N1 virus-specific and cross-reactive antibody and T cell responses in humans and animal models. Data from limited studies suggest that although initially robust, there is substantial waning of the serum antibody responses in survivors of H5N1 virus infection. Characterization of monoclonal antibodies generated from memory B cells of survivors of H5N1 virus infection has provided an understanding of the fine specificity of the human antibody response to H5N1 virus infection and identified strategies for immunotherapy. Human T cell responses induced by infection with seasonal influenza viruses are directed to relatively conserved internal proteins and cross-react with the H5N1 subtype. A role for T cell-based heterosubtypic immunity against H5N1 viruses is suggested in animal studies. Further studies on adaptive immune responses to H5N1 virus infection in both humans and animals are needed to inform the design of optimal immunological treatment and prevention modalities.
Collapse
Affiliation(s)
- Guus F Rimmelzwaan
- Viroscience Laboratory, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
46
|
Lee YN, Lee DH, Park JK, Yuk SS, Kwon JH, Nahm SS, Lee JB, Park SY, Choi IS, Song CS. Experimental infection and natural contact exposure of ferrets with canine influenza virus (H3N2). J Gen Virol 2013; 94:293-297. [PMID: 23329681 DOI: 10.1099/vir.0.042473-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Epidemics of H3N2 canine influenza virus (CIV) among dogs in South Korea and southern China have raised concern over the potential for zoonotic transmission of these viruses. Here, we analysed the pathogenesis and transmissibility of H3N2 CIV in ferret. H3N2 CIV replicated efficiently in the respiratory system of inoculated ferrets and caused acute necrotizing bronchioalveolitis and non-suppurative encephalitis. Transmission of H3N2 CIV was detected in three of six ferrets co-housed with inoculated ferrets, but no viruses were detected in second-contact ferrets. These findings show that H3N2 CIV has the capacity to replicate in and transmit partially among co-housed ferrets and underscore the need for continued public health surveillance.
Collapse
Affiliation(s)
- Yu-Na Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Dong-Hun Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Jae-Keun Park
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seong-Su Yuk
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Jung-Hoon Kwon
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Sang-Soep Nahm
- Laboratory of Veterinary Anatomy, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Joong-Bok Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seung-Yong Park
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - In-Soo Choi
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 143-701, Republic of Korea
| |
Collapse
|
47
|
Abstract
Respiratory viruses (including adenovirus, influenza virus, respiratory syncytial virus, coronavirus, and rhinovirus) cause a broad spectrum of disease in humans, ranging from mild influenza-like symptoms to acute respiratory failure. While species D adenoviruses and subtype H7 influenza viruses are known to possess an ocular tropism, documented human ocular disease has been reported following infection with all principal respiratory viruses. In this review, we describe the anatomical proximity and cellular receptor distribution between ocular and respiratory tissues. All major respiratory viruses and their association with human ocular disease are discussed. Research utilizing in vitro and in vivo models to study the ability of respiratory viruses to use the eye as a portal of entry as well as a primary site of virus replication is highlighted. Identification of shared receptor-binding preferences, host responses, and laboratory modeling protocols among these viruses provides a needed bridge between clinical and laboratory studies of virus tropism.
Collapse
Affiliation(s)
- Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | | | | |
Collapse
|
48
|
Plourde JR, Pyles JA, Layton RC, Vaughan SE, Tipper JL, Harrod KS. Neurovirulence of H5N1 infection in ferrets is mediated by multifocal replication in distinct permissive neuronal cell regions. PLoS One 2012; 7:e46605. [PMID: 23056366 PMCID: PMC3466300 DOI: 10.1371/journal.pone.0046605] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 09/03/2012] [Indexed: 12/31/2022] Open
Abstract
Highly pathogenic avian influenza A (HPAI), subtype H5N1, remains an emergent threat to the human population. While respiratory disease is a hallmark of influenza infection, H5N1 has a high incidence of neurological sequelae in many animal species and sporadically in humans. We elucidate the temporal/spatial infection of H5N1 in the brain of ferrets following a low dose, intranasal infection of two HPAI strains of varying neurovirulence and lethality. A/Vietnam/1203/2004 (VN1203) induced mortality in 100% of infected ferrets while A/Hong Kong/483/1997 (HK483) induced lethality in only 20% of ferrets, with death occurring significantly later following infection. Neurological signs were prominent in VN1203 infection, but not HK483, with seizures observed three days post challenge and torticollis or paresis at later time points. VN1203 and HK483 replication kinetics were similar in primary differentiated ferret nasal turbinate cells, and similar viral titers were measured in the nasal turbinates of infected ferrets. Pulmonary viral titers were not different between strains and pathological findings in the lungs were similar in severity. VN1203 replicated to high titers in the olfactory bulb, cerebral cortex, and brain stem; whereas HK483 was not recovered in these tissues. VN1203 was identified adjacent to and within the olfactory nerve tract, and multifocal infection was observed throughout the frontal cortex and cerebrum. VN1203 was also detected throughout the cerebellum, specifically in Purkinje cells and regions that coordinate voluntary movements. These findings suggest the increased lethality of VN1203 in ferrets is due to increased replication in brain regions important in higher order function and explains the neurological signs observed during H5N1 neurovirulence.
Collapse
Affiliation(s)
- Jennifer R. Plourde
- Infectious Diseases Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - John A. Pyles
- Infectious Diseases Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
| | - R. Colby Layton
- Infectious Diseases Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
| | - Sarah E. Vaughan
- Infectious Diseases Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Jennifer L. Tipper
- Infectious Diseases Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
| | - Kevin S. Harrod
- Infectious Diseases Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
49
|
Dengler L, May M, Wilk E, Bahgat MM, Schughart K. Immunization with live virus vaccine protects highly susceptible DBA/2J mice from lethal influenza A H1N1 infection. Virol J 2012; 9:212. [PMID: 22992381 PMCID: PMC3502422 DOI: 10.1186/1743-422x-9-212] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 09/13/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mouse represents an important model system to study the host response to influenza A infections and to evaluate new prevention or treatment strategies. We and others reported that the susceptibility to influenza A virus infections strongly varies among different inbred mouse strains. In particular, DBA/2J mice are highly susceptible to several influenza A subtypes, including human isolates and exhibit severe symptoms after infection with clinical isolates. FINDINGS Upon intra-muscular immunization with live H1N1 influenza A virus (mouse-adapted PR8M, and 2009 pandemic human HA04), DBA/2J mice mounted virus-specific IgG responses and were protected against a subsequent lethal challenge. The immune response and rescue from death after immunization in DBA/2J was similar to those observed for C57BL/6J mice. CONCLUSIONS DBA/2J mice represent a suitable mouse model to evaluate virulence and pathogenicity as well as immunization regimes against existing and newly emerging human influenza strains without the need for prior adaptation of the virus to the mouse.
Collapse
Affiliation(s)
- Leonie Dengler
- Department of Infection Genetics, Helmholtz Centre for Infection Research and University of Veterinary Medicine Hannover, Inhoffenstr. 7, D-38124 Braunschweig, Germany
| | | | | | | | | |
Collapse
|
50
|
Full genome sequence of a recombinant H5N1 influenza virus from a condor in southern China. J Virol 2012; 86:7722-3. [PMID: 22733885 DOI: 10.1128/jvi.01043-12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we report the first genomic information on an H5N1 avian influenza virus (AIV) isolated from a condor in Guangdong Province in southern China in 2003. Full genome sequencing and phylogenetic analyses show that it is a recombinant virus containing genome segments derived from the Eurasia and North America gene pools. This will be useful for analyses of the evolution of H5N1 AIV in southern China.
Collapse
|