1
|
Jin XY, Yang HY, Zhao GY, Dai CX, Zhang ZQ, Zhou DS, Yin Q, Dai EH. Comparative pathogenicity of influenza virus-induced pneumonia mouse model following intranasal and aerosolized intratracheal inoculation. Virol J 2024; 21:240. [PMID: 39354538 PMCID: PMC11446018 DOI: 10.1186/s12985-024-02516-6] [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: 06/27/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Infection of mice with mouse-adapted strains of influenza virus has been widely used to establish mouse pneumonia models. Intranasal inoculation is the traditional route for constructing an influenza virus-induced pneumonia mouse model, while intratracheal inoculation has been gradually applied in recent years. In this article, the pathogenicity of influenza virus-induced pneumonia mouse models following intranasal and aerosolized intratracheal inoculation were compared. METHODS By comparing the two ways of influenza inoculation, intranasal and intratracheal, a variety of indices such as survival rate, body weight change, viral titer and load, pathological change, lung wet/dry ratio, and inflammatory factors were investigated. Meanwhile, the transcriptome was applied for the initial exploration of the mechanism underlying the variations in the results between the two inoculation methods. RESULTS The findings suggest that aerosolized intratracheal infection leads to more severe lung injury and higher viral loads in the lungs compared to intranasal infection, which may be influenced by the initial site of infection, sialic acid receptor distribution, and host innate immunity. CONCLUSION Intratracheal inoculation is a better method for modelling severe pneumonia in mice than intranasal infection.
Collapse
Affiliation(s)
- Xiu-Yu Jin
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, People's Republic of China
| | - Hui-Ying Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Guang-Yu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Chen-Xi Dai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Zai-Qing Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Dong-Sheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Er-Hei Dai
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, People's Republic of China.
| |
Collapse
|
2
|
Varying Viral Replication and Disease Profiles of H2N2 Influenza in Ferrets Is Associated with Virus Isolate and Inoculation Route. J Virol 2022; 96:e0073222. [PMID: 35862678 PMCID: PMC9327684 DOI: 10.1128/jvi.00732-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In 1957 the world was subjected to a pandemic caused by an influenza A virus of the subtype H2N2. Although the virus disappeared in 1968, H2 viruses continue to circulate in avian reservoirs.
Collapse
|
3
|
Bi Z, Hong W, Yang J, Lu S, Peng X. Animal models for SARS-CoV-2 infection and pathology. MedComm (Beijing) 2021; 2:548-568. [PMID: 34909757 PMCID: PMC8662225 DOI: 10.1002/mco2.98] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiology of coronavirus disease 2019 (COVID-19) pandemic. Current variants including Alpha, Beta, Gamma, Delta, and Lambda increase the capacity of infection and transmission of SARS-CoV-2, which might disable the in-used therapies and vaccines. The COVID-19 has now put an enormous strain on health care system all over the world. Therefore, the development of animal models that can capture characteristics and immune responses observed in COVID-19 patients is urgently needed. Appropriate models could accelerate the testing of therapeutic drugs and vaccines against SARS-CoV-2. In this review, we aim to summarize the current animal models for SARS-CoV-2 infection, including mice, hamsters, nonhuman primates, and ferrets, and discuss the details of transmission, pathology, and immunology induced by SARS-CoV-2 in these animal models. We hope this could throw light to the increased usefulness in fundamental studies of COVID-19 and the preclinical analysis of vaccines and therapeutic agents.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Shuaiyao Lu
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| | - Xiaozhong Peng
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| |
Collapse
|
4
|
van de Ven K, van Dijken H, Wijsman L, Gomersbach A, Schouten T, Kool J, Lenz S, Roholl P, Meijer A, van Kasteren PB, de Jonge J. Pathology and Immunity After SARS-CoV-2 Infection in Male Ferrets Is Affected by Age and Inoculation Route. Front Immunol 2021; 12:750229. [PMID: 34745122 PMCID: PMC8566349 DOI: 10.3389/fimmu.2021.750229] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Improving COVID-19 intervention strategies partly relies on animal models to study SARS-CoV-2 disease and immunity. In our pursuit to establish a model for severe COVID-19, we inoculated young and adult male ferrets intranasally or intratracheally with SARS-CoV-2. Intranasal inoculation established an infection in all ferrets, with viral dissemination into the brain and gut. Upon intratracheal inoculation only adult ferrets became infected. However, neither inoculation route induced observable COVID-19 symptoms. Despite this, a persistent inflammation in the nasal turbinates was prominent in especially young ferrets and follicular hyperplasia in the bronchi developed 21 days post infection. These effects -if sustained- might resemble long-COVID. Respiratory and systemic cellular responses and antibody responses were induced only in animals with an established infection. We conclude that intranasally-infected ferrets resemble asymptomatic COVID-19 and possibly aspects of long-COVID. Combined with the increasing portfolio to measure adaptive immunity, ferrets are a relevant model for SARS-CoV-2 vaccine research.
Collapse
Affiliation(s)
- Koen van de Ven
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | - Harry van Dijken
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | - Lisa Wijsman
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | - Angéla Gomersbach
- Animal Research Centre, Poonawalla Science Park, Bilthoven, Netherlands
| | - Tanja Schouten
- Animal Research Centre, Poonawalla Science Park, Bilthoven, Netherlands
| | - Jolanda Kool
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | - Stefanie Lenz
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | | | - Adam Meijer
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | - Puck B van Kasteren
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| | - Jørgen de Jonge
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
5
|
Stadlbauer D, Waal LD, Beaulieu E, Strohmeier S, Kroeze EJBV, Boutet P, Osterhaus ADME, Krammer F, Innis BL, Nachbagauer R, Stittelaar KJ, Mallett CP. AS03-adjuvanted H7N9 inactivated split virion vaccines induce cross-reactive and protective responses in ferrets. NPJ Vaccines 2021; 6:40. [PMID: 33742000 PMCID: PMC7979725 DOI: 10.1038/s41541-021-00299-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 02/16/2021] [Indexed: 01/09/2023] Open
Abstract
Human infections with avian H7N9 subtype influenza viruses are a major public health concern and vaccines against H7N9 are urgently needed for pandemic preparedness. In early 2013, novel H7N9 influenza viruses emerged in China that caused about 1600 human cases of infection with a high associated case fatality rate. In this study, two H7N9 split virion vaccines with or without AS03 adjuvant were tested in the naive ferret model. Serological analyses demonstrated that homologous hemagglutination inhibition and microneutralization antibody titers were detectable in the ferrets after the first immunization with the AS03-adjuvanted vaccines that were further boosted by the second immunization. In addition, heterologous antibody titers against older H7 subtype viruses of the North American lineage (H7N7, H7N3) and newer H7 subtype viruses of the Eurasian lineage (H7N9) were detected in the animals receiving the AS03-adjuvanted vaccines. Animals receiving two immunizations of the AS03-adjuvanted vaccines were protected from weight loss and fever in the homologous challenge study and had no detectable virus in throat or lung samples. In addition, microscopic examination post-challenge showed animals immunized with the AS03-adjuvanted vaccines had the least signs of lung injury and inflammation, consistent with the greater relative efficacy of the adjuvanted vaccines. In conclusion, this study demonstrated that the AS03-adjuvanted H7N9 vaccines elicited high levels of homologous and heterologous antibodies and protected against H7N9 virus damage post-challenge.
Collapse
Affiliation(s)
- Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leon de Waal
- Viroclinics Biosciences B.V., Viroclinics Xplore, Schaijk, The Netherlands
| | | | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | | | | | - Albert D M E Osterhaus
- Viroclinics Biosciences B.V., Viroclinics Xplore, Schaijk, The Netherlands.,The Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce L Innis
- GSK, King of Prussia, PA, USA.,PATH, Center for Vaccine Innovation and Access, Washington, DC, USA
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Moderna Inc., Cambridge, MA, USA
| | - Koert J Stittelaar
- Viroclinics Biosciences B.V., Viroclinics Xplore, Schaijk, The Netherlands.,Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | | |
Collapse
|
6
|
Sialic Acid Receptors: The Key to Solving the Enigma of Zoonotic Virus Spillover. Viruses 2021; 13:v13020262. [PMID: 33567791 PMCID: PMC7915228 DOI: 10.3390/v13020262] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Emerging viral diseases are a major threat to global health, and nearly two-thirds of emerging human infectious diseases are zoonotic. Most of the human epidemics and pandemics were caused by the spillover of viruses from wild mammals. Viruses that infect humans and a wide range of animals have historically caused devastating epidemics and pandemics. An in-depth understanding of the mechanisms of viral emergence and zoonotic spillover is still lacking. Receptors are major determinants of host susceptibility to viruses. Animal species sharing host cell receptors that support the binding of multiple viruses can play a key role in virus spillover and the emergence of novel viruses and their variants. Sialic acids (SAs), which are linked to glycoproteins and ganglioside serve as receptors for several human and animal viruses. In particular, influenza and coronaviruses, which represent two of the most important zoonotic threats, use SAs as cellular entry receptors. This is a comprehensive review of our current knowledge of SA receptor distribution among animal species and the range of viruses that use SAs as receptors. SA receptor tropism and the predicted natural susceptibility to viruses can inform targeted surveillance of domestic and wild animals to prevent the future emergence of zoonotic viruses.
Collapse
|
7
|
Adami EA, Chavez Rico SL, Akamatsu MA, Miyaki C, Raw I, de Oliveira D, Comone P, Oliveira RDN, Sarno de Oliveira ML, Estima Abreu PA, Takano CY, Meros M, Soares-Schanoski A, Lee Ho P. H7N9 pandemic preparedness: A large-scale production of a split inactivated vaccine. Biochem Biophys Res Commun 2021; 545:145-149. [PMID: 33550095 DOI: 10.1016/j.bbrc.2021.01.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/19/2021] [Indexed: 11/18/2022]
Abstract
In March 2013 it was reported by the World Health Organization (WHO) the first cases of human infections with avian influenza virus A (H7N9). From 2013 to December 2019, 1568 cases have been reported with 616 deaths. H7N9 infection has been associated with high morbidity and mortality rates, and vaccination is currently the most effective way to prevent infections and consequently flu-related severe illness. Developing and producing vaccines against pandemic influenza viruses is the main strategy for a response to a possible pandemic. This study aims to present the production of three industrial lots under current Good Manufacturing Practices (cGMP) of the active antigen used to produce the pandemic influenza vaccine candidate against A(H7N9). These batches were characterized and evaluated for quality standards and tested for immunogenicity in mice. The average yield was 173.50 ± 7.88 μg/mL of hemagglutinin and all the preparations met all the required specifications. The formulated H7N9 vaccine is poorly immunogenic and needs to be adjuvanted with an oil in water emulsion adjuvant (IB160) to achieve a best immune response, in a prime and in a boost scheme. These data are important for initial production planning and preparedness in the case of a H7N9 pandemic.
Collapse
MESH Headings
- Animals
- Antigens, Viral/biosynthesis
- Antigens, Viral/immunology
- Drug Compounding/methods
- Drug Compounding/statistics & numerical data
- Drug Industry/standards
- Female
- Humans
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza Vaccines/biosynthesis
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Pandemics/prevention & control
- Vaccines, Inactivated/biosynthesis
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/isolation & purification
Collapse
Affiliation(s)
| | | | | | | | - Isaías Raw
- Biotechnology Center, Butantan Institute, 05503-900, SP, Brazil
| | | | | | | | | | | | | | | | - Alessandra Soares-Schanoski
- Bacteriology Laboratory, Butantan Institute, Brazil; Icahn School of Medicine at Mount Sinai, New York, USA.
| | | |
Collapse
|
8
|
H7N9 influenza split vaccine with SWE oil-in-water adjuvant greatly enhances cross-reactive humoral immunity and protection against severe pneumonia in ferrets. NPJ Vaccines 2020; 5:38. [PMID: 32411401 PMCID: PMC7214439 DOI: 10.1038/s41541-020-0187-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 04/21/2020] [Indexed: 12/24/2022] Open
Abstract
Until universal influenza vaccines become available, pandemic preparedness should include developing classical vaccines against potential pandemic influenza subtypes. We here show that addition of SWE adjuvant, a squalene-in-water emulsion, to H7N9 split influenza vaccine clearly enhanced functional antibody responses in ferrets. These were cross-reactive against H7N9 strains from different lineages and newly emerged H7N9 variants. Both vaccine formulations protected in almost all cases against severe pneumonia induced by intratracheal infection of ferrets with H7N9 influenza; however, the SWE adjuvant enhanced protection against virus replication and disease. Correlation analysis and curve fitting showed that both VN- and NI-titers were better predictors for protection than HI-titers. Moreover, we show that novel algorithms can assist in better interpretation of large data sets generated in preclinical studies. Cluster analysis showed that the adjuvanted vaccine results in robust immunity and protection, whereas the response to the non-adjuvanted vaccine is heterogeneous, such that the protection balance may be more easily tipped toward severe disease. Finally, cluster analysis indicated that the dose-sparing capacity of the adjuvant is at least a factor six, which greatly increases vaccine availability in a pandemic situation.
Collapse
|
9
|
Elbers ARW, Gonzales JL. Quantification of visits of wild fauna to a commercial free-range layer farm in the Netherlands located in an avian influenza hot-spot area assessed by video-camera monitoring. Transbound Emerg Dis 2020; 67:661-677. [PMID: 31587498 PMCID: PMC7079184 DOI: 10.1111/tbed.13382] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 09/11/2019] [Accepted: 09/22/2019] [Indexed: 01/19/2023]
Abstract
Free-range poultry farms have a high risk of introduction of avian influenza viruses (AIV), and it is presumed that wild (water) birds are the source of introduction. There is very scarce quantitative data on wild fauna visiting free-range poultry farms. We quantified visits of wild fauna to a free-range area of a layer farm, situated in an AIV hot-spot area, assessed by video-camera monitoring. A total of 5,016 hr (209 days) of video recordings, covering all 12 months of a year, were analysed. A total of 16 families of wild birds and five families of mammals visited the free-range area of the layer farm. Wild birds, except for the dabbling ducks, visited the free-range area almost exclusively in the period between sunrise and the moment the chickens entered the free-range area. Known carriers of AIV visited the outdoor facility regularly: species of gulls almost daily in the period January-August; dabbling ducks only in the night in the period November-May, with a distinct peak in the period December-February. Only a small fraction of visits of wild fauna had overlap with the presence of chickens at the same time in the free-range area. No direct contact between chickens and wild birds was observed. It is hypothesized that AIV transmission to poultry on free-range poultry farms will predominantly take place via indirect contact: taking up AIV by chickens via wild-bird-faeces-contaminated water or soil in the free-range area. The free-range poultry farmer has several possibilities to potentially lower the attractiveness of the free-range area for wild (bird) fauna: daily inspection of the free-range area and removal of carcasses and eggs; prevention of forming of water pools in the free-range facility. Furthermore, there are ways to scare-off wild birds, for example use of laser equipment or trained dogs.
Collapse
Affiliation(s)
- Armin R. W. Elbers
- Department of Bacteriology and EpidemiologyWageningen Bioveterinary ResearchLelystadThe Netherlands
| | - José L. Gonzales
- Department of Bacteriology and EpidemiologyWageningen Bioveterinary ResearchLelystadThe Netherlands
| |
Collapse
|
10
|
Wang WH, Erazo EM, Ishcol MRC, Lin CY, Assavalapsakul W, Thitithanyanont A, Wang SF. Virus-induced pathogenesis, vaccine development, and diagnosis of novel H7N9 avian influenza A virus in humans: a systemic literature review. J Int Med Res 2019; 48:300060519845488. [PMID: 31068040 PMCID: PMC7140199 DOI: 10.1177/0300060519845488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
H7N9 avian influenza virus (AIV) caused human infections in 2013 in China.
Phylogenetic analyses indicate that H7N9 AIV is a novel reassortant strain with
pandemic potential. We conducted a systemic review regarding virus-induced
pathogenesis, vaccine development, and diagnosis of H7N9 AIV infection in
humans. We followed PRISMA guidelines and searched PubMed, Web of Science, and
Google Scholar to identify relevant articles published between January 2013 and
December 2018. Pathogenesis data indicated that H7N9 AIV belongs to low
pathogenic avian influenza, which is mostly asymptomatic in avian species;
however, H7N9 induces high mortality in humans. Sporadic human infections have
recently been reported, caused by highly pathogenic avian influenza viruses
detected in poultry. H7N9 AIVs resistant to adamantine and oseltamivir cause
severe human infection by rapidly inducing progressive acute community-acquired
pneumonia, multiorgan dysfunction, and cytokine dysregulation; however,
mechanisms via which the virus induces severe syndromes remain unclear. An H7N9
AIV vaccine is lacking; designs under evaluation include synthesized peptide,
baculovirus-insect system, and virus-like particle vaccines. Molecular diagnosis
of H7N9 AIVs is suggested over conventional assays, for biosafety reasons.
Several advanced or modified diagnostic assays are under investigation and
development. We summarized virus-induced pathogenesis, vaccine development, and
current diagnostic assays in H7N9 AIVs.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung.,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung
| | - Esmeralda Merari Erazo
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung
| | - Max R Chang Ishcol
- Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung
| | - Chih-Yen Lin
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Sheng-Fan Wang
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung
| |
Collapse
|
11
|
Influenza A Virus Reassortment Is Limited by Anatomical Compartmentalization following Coinfection via Distinct Routes. J Virol 2018; 92:JVI.02063-17. [PMID: 29212934 DOI: 10.1128/jvi.02063-17] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/22/2022] Open
Abstract
Exchange of gene segments through reassortment is a major feature of influenza A virus evolution and frequently contributes to the emergence of novel epidemic, pandemic, and zoonotic strains. It has long been evident that viral diversification through reassortment is constrained by genetic incompatibility between divergent parental viruses. In contrast, the role of virus-extrinsic factors in determining the likelihood of reassortment has remained unclear. To evaluate the impact of such factors in the absence of confounding effects of segment mismatch, we previously reported an approach in which reassortment between wild-type (wt) and genetically tagged variant (var) viruses of the same strain is measured. Here, using wt/var systems in the A/Netherlands/602/2009 (pH1N1) and A/Panama/2007/99 (H3N2) strain backgrounds, we tested whether inoculation of parental viruses into distinct sites within the respiratory tract limits their reassortment. Using a ferret (Mustella putorius furo) model, either matched parental viruses were coinoculated intranasally or one virus was instilled intranasally whereas the second was instilled intratracheally. Dual intranasal inoculation resulted in robust reassortment for wt/var viruses of both strain backgrounds. In contrast, when infections were initiated simultaneously at distinct sites, strong compartmentalization of viral replication was observed and minimal reassortment was detected. The observed lack of viral spread between upper and lower respiratory tract tissues may be attributable to localized exclusion of superinfection within the host, mediated by innate immune responses. Our findings indicate that dual infections in nature are more likely to result in reassortment if viruses are seeded into similar anatomical locations and have matched tissue tropisms.IMPORTANCE Genetic exchange between influenza A viruses (IAVs) through reassortment can facilitate the emergence of antigenically drifted seasonal strains and plays a prominent role in the development of pandemics. Typical human influenza infections are concentrated in the upper respiratory tract; however, lower respiratory tract (LRT) infection is an important feature of severe cases, which are more common in the very young, the elderly, and individuals with underlying conditions. In addition to host factors, viral characteristics and mode of transmission can also increase the likelihood of LRT infection: certain zoonotic IAVs are thought to favor the LRT, and transmission via small droplets allows direct seeding into lower respiratory tract tissues. To gauge the likelihood of reassortment in coinfected hosts, we assessed the extent to which initiation of infection at distinct respiratory tract sites impacts reassortment frequency. Our results reveal that spatially distinct inoculations result in anatomical compartmentalization of infection, which in turn strongly limits reassortment.
Collapse
|
12
|
Liu L, Lu J, Li Z, Zhou J, Guo J, Li X, Liu J, Shu Y, Wang D. 220 mutation in the hemagglutinin of avian influenza A (H7N9) virus alters antigenicity during vaccine strain development. Hum Vaccin Immunother 2018; 14:532-539. [PMID: 29300686 PMCID: PMC5861781 DOI: 10.1080/21645515.2017.1419109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
Since the first confirmed case of H7N9 infection was reported in China, there have been five epidemic waves of human H7N9 infections between 2013 and 2017. The fifth wave differed from the previous four waves in that highly pathogenic avian influenza (HPAI) H7N9 viruses with multiple basic amino acids at the cleavage site were detected in humans, poultry and environmental samples. The HPAI H7N9 viruses were genetically and antigenically distinct from previous H7N9 viruses. Therefore, a new candidate vaccine virus(CVV) derived from a HPAI A/Guangdong/17SF003/2016-like virus was proposed by the World Health Organization(WHO). According to the WHO recommendations, we constructed a new CVV using reverse genetic technology, with a (6+2) gene constitution. The (6+2) reassortant virus possessed hemagglutinin(HA) with multiple basic amino acids removed and the neuraminidase from A/Guangdong/SF003/2016 in a high-yield A/Puerto Rico/8/34 virus backbone. Sequence analysis confirmed that no mutations had occurred in the HA of V1E1(the initial CVV rescued in Vero cells and followed by passage in eggs), but a mixture of arginine (R)/glycine (G)/isoleucine (I) was detected at position 220 (H3 numbering) in the HA of V1E2 to V1E5 with different percentages. Furthermore, V1E5 showed improved growth characteristics and immunogenicity compared with V1E1, and retained low pathogenicity in chickens and chicken embryos, but the mutation changed its antigenicity. Our study indicates that antigenic changes should be closely monitored during the development of H7N9 CVV in eggs. Additionally, although V1E5 changes the antigenicity, the antisera had some reactivity to previous H7N9 CVVs.
Collapse
Affiliation(s)
- Liqi Liu
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Jian Lu
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Zi Li
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Jianfang Zhou
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Junfeng Guo
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Xiyan Li
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Jia Liu
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| | - Yuelong Shu
- b Public Health School(Shenzhen), Sun Yat-sen University , P. R. China
| | - Dayan Wang
- a Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention , Beijing , China CDC
| |
Collapse
|
13
|
Chang YF, Wang WH, Hong YW, Yuan RY, Chen KH, Huang YW, Lu PL, Chen YH, Chen YMA, Su LC, Wang SF. Simple Strategy for Rapid and Sensitive Detection of Avian Influenza A H7N9 Virus Based on Intensity-Modulated SPR Biosensor and New Generated Antibody. Anal Chem 2018; 90:1861-1869. [PMID: 29327590 DOI: 10.1021/acs.analchem.7b03934] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In 2013 a new reassortant avian influenza A H7N9 virus emerged in China, causing human infection with high mortality. An accurate and timely diagnosis is crucial for controlling the outbreaks of the disease. We therefore propose a simple strategy for rapidly and sensitively detecting the H7N9 virus using an intensity-modulated surface plasmon resonance (IM-SPR) biosensor integrated with a new generated monoclonal antibody. The novel antibody exhibits significant specificity to recognize H7N9 virus compared with other clinical human influenza isolates (p < 0.01). Experimentally, the detection limit of the proposed approach for H7N9 virus detection is estimated to be 144 copies/mL, which is a 20-fold increase in sensitivity compared with homemade target-captured ELISA using the identical antibody. For the measurement of mimic clinical specimens containing the H7N9 virus mixed with nasal mucosa from flu-like syndrome patients, the detection limit is calculated to be 402 copies/mL, which is better than conventional influenza detection assays; quantitative reverse transcription polymerase chain reaction (qRT-PCR) and rapid influenza diagnostic test (RIDT). Most importantly, the assay time took less than 10 min. Combined, the results of this study indicate that the proposed simple strategy demonstrates high sensitivity and time-saving in H7N9 virus detection. By incorporating a high specific recognizer, the proposed technique has the potential to be used in applications and development of other emerging or re-emerging microbe detection platforms.
Collapse
Affiliation(s)
- Ying-Feng Chang
- Bio-Analytical Chemistry and Nanobiomedicine Laboratory, Department of Biochemical Science and Technology, National Taiwan University , Taipei 10617, Taiwan
| | - Wen-Hung Wang
- Division of Infection Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital , Kaohsiung 80708, Taiwan.,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Yi-Wei Hong
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University , Kaohsiung 80708, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Ruei-Yu Yuan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University , Kaohsiung 80708, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Kuan-Hsuan Chen
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Yu-Wen Huang
- Dermatology department, Yuan's general hospital , Kaohsiung 80249, Taiwan
| | - Po-Liang Lu
- Division of Infection Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital , Kaohsiung 80708, Taiwan.,Department of Laboratory Medicine, Kaohsiung Medical University Hospital , Kaohsiung 80708, Taiwan
| | - Yen-Hsu Chen
- Division of Infection Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital , Kaohsiung 80708, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Yi-Ming Arthur Chen
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University , Kaohsiung 80708, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Li-Chen Su
- Department of Optoelectric Physics, Chinese Culture University , Taipei 11114, Taiwan
| | - Sheng-Fan Wang
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University , Kaohsiung 80708, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University , Kaohsiung 80708, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital , Kaohsiung 81267, Taiwan
| |
Collapse
|
14
|
Velkers FC, Blokhuis SJ, Veldhuis Kroeze EJB, Burt SA. The role of rodents in avian influenza outbreaks in poultry farms: a review. Vet Q 2017; 37:182-194. [DOI: 10.1080/01652176.2017.1325537] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Francisca C. Velkers
- Department of Farm Animal Health – Epidemiology, Infectiology and Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Simon J. Blokhuis
- Department of Farm Animal Health – Epidemiology, Infectiology and Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | - Sara A. Burt
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
15
|
de Jonge J, Isakova-Sivak I, van Dijken H, Spijkers S, Mouthaan J, de Jong R, Smolonogina T, Roholl P, Rudenko L. H7N9 Live Attenuated Influenza Vaccine Is Highly Immunogenic, Prevents Virus Replication, and Protects Against Severe Bronchopneumonia in Ferrets. Mol Ther 2016; 24:991-1002. [PMID: 26796670 PMCID: PMC4881767 DOI: 10.1038/mt.2016.23] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/22/2015] [Indexed: 12/19/2022] Open
Abstract
Avian influenza viruses continue to cross the species barrier, and if such viruses become transmissible among humans, it would pose a great threat to public health. Since its emergence in China in 2013, H7N9 has caused considerable morbidity and mortality. In the absence of a universal influenza vaccine, preparedness includes development of subtype-specific vaccines. In this study, we developed and evaluated in ferrets an intranasal live attenuated influenza vaccine (LAIV) against H7N9 based on the A/Leningrad/134/17/57 (H2N2) cold-adapted master donor virus. We demonstrate that the LAIV is attenuated and safe in ferrets and induces high hemagglutination- and neuraminidase-inhibiting and virus-neutralizing titers. The antibodies against hemagglutinin were also cross-reactive with divergent H7 strains. To assess efficacy, we used an intratracheal challenge ferret model in which an acute severe viral pneumonia is induced that closely resembles viral pneumonia observed in severe human cases. A single- and two-dose strategy provided complete protection against severe pneumonia and prevented virus replication. The protective effect of the two-dose strategy appeared better than the single dose only on the microscopic level in the lungs. We observed, however, an increased lymphocytic infiltration after challenge in single-vaccinated animals and hypothesize that this a side effect of the model.
Collapse
Affiliation(s)
- Jørgen de Jonge
- Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Harry van Dijken
- Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Sanne Spijkers
- Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, the Netherlands
- Current address: BioNovion, Oss, the Netherlands
| | - Justin Mouthaan
- Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, the Netherlands
- Current address: Genmab, Utrecht, the Netherlands
| | - Rineke de Jong
- Department of Virology, Central Veterinary Institute of Wageningen UR, Lelystad, the Netherlands
| | - Tatiana Smolonogina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Paul Roholl
- Microscope Consultancy, Weesp, the Netherlands
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| |
Collapse
|
16
|
Oh DY, Hurt AC. Using the Ferret as an Animal Model for Investigating Influenza Antiviral Effectiveness. Front Microbiol 2016; 7:80. [PMID: 26870031 PMCID: PMC4740393 DOI: 10.3389/fmicb.2016.00080] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/18/2016] [Indexed: 01/12/2023] Open
Abstract
The concern of the emergence of a pandemic influenza virus has sparked an increased effort toward the development and testing of novel influenza antivirals. Central to this is the animal model of influenza infection, which has played an important role in understanding treatment effectiveness and the effect of antivirals on host immune responses. Among the different animal models of influenza, ferrets can be considered the most suitable for antiviral studies as they display most of the human-like symptoms following influenza infections, they can be infected with human influenza virus without prior viral adaptation and have the ability to transmit influenza virus efficiently between one another. However, an accurate assessment of the effectiveness of an antiviral treatment in ferrets is dependent on three major experimental considerations encompassing firstly, the volume and titer of virus, and the route of viral inoculation. Secondly, the route and dose of drug administration, and lastly, the different methods used to assess clinical symptoms, viral shedding kinetics and host immune responses in the ferrets. A good understanding of these areas is necessary to achieve data that can accurately inform the human use of influenza antivirals. In this review, we discuss the current progress and the challenges faced in these three major areas when using the ferret model to measure influenza antiviral effectiveness.
Collapse
Affiliation(s)
- Ding Y Oh
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, MelbourneVIC, Australia; School of Applied and Biomedical Sciences, Federation University Australia, GippslandVIC, Australia
| | - Aeron C Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, MelbourneVIC, Australia; Melbourne School of Population and Global Health, University of Melbourne, ParkvilleVIC, Australia
| |
Collapse
|
17
|
Renukaradhya GJ, Narasimhan B, Mallapragada SK. Respiratory nanoparticle-based vaccines and challenges associated with animal models and translation. J Control Release 2015; 219:622-631. [PMID: 26410807 PMCID: PMC4760633 DOI: 10.1016/j.jconrel.2015.09.047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022]
Abstract
Vaccine development has had a huge impact on human health. However, there is a significant need to develop efficacious vaccines for several existing as well as emerging respiratory infectious diseases. Several challenges need to be overcome to develop efficacious vaccines with translational potential. This review focuses on two aspects to overcome some barriers - 1) the development of nanoparticle-based vaccines, and 2) the choice of suitable animal models for respiratory infectious diseases that will allow for translation. Nanoparticle-based vaccines, including subunit vaccines involving synthetic and/or natural polymeric adjuvants and carriers, as well as those based on virus-like particles offer several key advantages to help overcome the barriers to effective vaccine development. These include the ability to deliver combinations of antigens, target the vaccine formulation to specific immune cells, enable cross-protection against divergent strains, act as adjuvants or immunomodulators, allow for sustained release of antigen, enable single dose delivery, and potentially obviate the cold chain. While mouse models have provided several important insights into the mechanisms of infectious diseases, they are often a limiting step in translation of new vaccines to the clinic. An overview of different animal models involved in vaccine research for respiratory infections, with advantages and disadvantages of each model, is discussed. Taken together, advances in nanotechnology, combined with the right animal models for evaluating vaccine efficacy, has the potential to revolutionize vaccine development for respiratory infections.
Collapse
Affiliation(s)
- Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, United States
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
18
|
Abstract
Transmission via shared water implicates passerine birds as possible vectors for dissemination of this virus. Low pathogenicity avian influenza A(H7N9) virus has been detected in poultry since 2013, and the virus has caused >450 infections in humans. The mode of subtype H7N9 virus transmission between avian species remains largely unknown, but various wild birds have been implicated as a source of transmission. H7N9 virus was recently detected in a wild sparrow in Shanghai, China, and passerine birds, such as finches, which share space and resources with wild migratory birds, poultry, and humans, can be productively infected with the virus. We demonstrate that interspecies transmission of H7N9 virus occurs readily between society finches and bobwhite quail but only sporadically between finches and chickens. Inoculated finches are better able to infect naive poultry than the reverse. Transmission occurs through shared water but not through the airborne route. It is therefore conceivable that passerine birds may serve as vectors for dissemination of H7N9 virus to domestic poultry.
Collapse
|
19
|
Root JJ, Bosco-Lauth AM, Bielefeldt-Ohmann H, Bowen RA. Experimental infection of peridomestic mammals with emergent H7N9 (A/Anhui/1/2013) influenza A virus: Implications for biosecurity and wet markets. Virology 2015; 487:242-8. [PMID: 26550948 PMCID: PMC7127772 DOI: 10.1016/j.virol.2015.10.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/10/2015] [Accepted: 10/19/2015] [Indexed: 11/26/2022]
Abstract
During 2013, a novel avian-origin H7N9 influenza A virus (IAV) emerged in China and subsequently caused large economic and public health burdens. We experimentally infected three common peridomestic wild mammals with H7N9 (A/Anhui/1/2013) IAV. Striped skunks exhibited the highest burden of disease followed by raccoons and cottontail rabbits. Striped skunks also produced the highest levels of viral shedding (up to 10(6.4)PFU/mL nasal flush) followed by cottontail rabbits (up to 10(5.8)PFU/mL nasal flush) and raccoons (up to 10(5.2)PFU/mL nasal flush). Thus, various mammalian species, especially those that are peridomestic, could play a role in the epidemiology of emergent H7N9 IAV. Mammals should be accounted for in biosecurity plans associated with H7N9 and their presence in wet markets, dependent on species, could lead to increased transmission among interspecific species aggregations and may also pose an elevated zoonotic disease risk to visitors and workers of such markets.
Collapse
Affiliation(s)
- J Jeffrey Root
- United States Department of Agriculture, Fort Collins, CO 80521, USA.
| | | | | | | |
Collapse
|
20
|
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
|
21
|
Virus replication kinetics and pathogenesis of infection with H7N9 influenza virus in isogenic guinea pigs upon intratracheal inoculation. Vaccine 2015; 33:6983-7. [PMID: 26307235 DOI: 10.1016/j.vaccine.2015.08.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 11/20/2022]
Abstract
Since 2013, avian influenza viruses of subtype H7N9 have been transmitted from poultry to humans in China and caused severe disease. Concerns persist over the pandemic potential of this virus and further understanding of immunity and transmission is required. The isogenic guinea pig model uniquely would allow for investigation into both. Eighteen female isogenic guinea pigs 12-16 weeks were inoculated intratracheally with either A/H7N9 virus (n=12) or PBS (n=6) and sacrificed on days 2 and 7 post-inoculation. Nasal and pharyngeal swabs were taken daily to assess viral replication kinetics and necropsies were performed to study pathogenesis. All animals showed peak virus titers in nasal secretions at day 2 post-inoculation and by day 7 post-inoculation infectious virus titers had decreased to just above detectable levels. At day 2, high virus titers were found in nasal turbinates and lungs and moderate titers in trachea and cerebrum. At day 7, infectious virus was detected in the nasal turbinates only. Histology showed moderate to severe inflammation in the entire respiratory tract and immunohistochemistry (IHC) demonstrated large numbers of viral antigen positive cells in the nasal epithelium at day 2 and fewer at day 7 post-inoculation. A moderate number of IHC positive cells was observed in the bronchi(oli) and alveoli at day 2 only. This study indicates that isogenic guinea pigs are a promising model to further study immunity to and transmission of H7N9 influenza virus.
Collapse
|
22
|
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: 121] [Impact Index Per Article: 13.4] [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
|
23
|
Pathogenesis of novel reassortant avian influenza virus A (H5N8) Isolates in the ferret. Virology 2015; 481:136-41. [PMID: 25776760 DOI: 10.1016/j.virol.2015.02.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/15/2014] [Accepted: 02/20/2015] [Indexed: 11/20/2022]
Abstract
Outbreaks of avian influenza virus H5N8 first occurred in 2014, and spread to poultry farms in Korea. Although there was no report of human infection by this subtype, it has the potential to threaten human public health. Therefore, we evaluated the pathogenesis of H5N8 viruses in ferrets. Two representative Korean H5N8 strains did not induce mortality and significant respiratory signs after an intranasal challenge in ferrets. However, ferrets intratracheally infected with A/broiler duck/Korea/Buan2/2014 virus showed dose-dependent mortality. Although the Korean H5N8 strains were classified as the HPAI virus, possessing multiple basic amino acids in the cleavage site of the hemagglutinin sequence, they did not produce pathogenesis in ferrets challenged intranasally, similar to the natural infection route. These results could be useful for public health by providing the pathogenic characterization of H5N8 viruses.
Collapse
|
24
|
van Dijk JGB, Kleyheeg E, Soons MB, Nolet BA, Fouchier RAM, Klaassen M. Weak negative associations between avian influenza virus infection and movement behaviour in a key host species, the mallard
Anas platyrhynchos. OIKOS 2015. [DOI: 10.1111/oik.01836] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Jacintha G. B. van Dijk
- Dept of Animal Ecology Netherlands Inst. of Ecology (NIOO‐KNAW) Droevendaalsesteeg 10 NL‐6708 PB Wageningen the Netherlands
| | - Erik Kleyheeg
- Inst. of Environmental Biology, Utrecht Univ. Padualaan 8 NL‐3584 CH Utrecht the Netherlands
| | - Merel B. Soons
- Dept of Animal Ecology Netherlands Inst. of Ecology (NIOO‐KNAW) Droevendaalsesteeg 10 NL‐6708 PB Wageningen the Netherlands
- Inst. of Environmental Biology, Utrecht Univ. Padualaan 8 NL‐3584 CH Utrecht the Netherlands
| | - Bart A. Nolet
- Dept of Animal Ecology Netherlands Inst. of Ecology (NIOO‐KNAW) Droevendaalsesteeg 10 NL‐6708 PB Wageningen the Netherlands
| | - Ron A. M. Fouchier
- Dept of Viroscience Erasmus MC PO Box 2040, NL‐3000 CA Rotterdam the Netherlands
| | - Marcel Klaassen
- Dept of Animal Ecology Netherlands Inst. of Ecology (NIOO‐KNAW) Droevendaalsesteeg 10 NL‐6708 PB Wageningen the Netherlands
- Centre for Integrative Ecology, School of Life and Environmental Sciences, Deakin Univ. Locked Bag 20000 Geelong VIC 3220 Australia
| |
Collapse
|
25
|
van Dijk JGB, Fouchier RAM, Klaassen M, Matson KD. Minor differences in body condition and immune status between avian influenza virus-infected and noninfected mallards: a sign of coevolution? Ecol Evol 2014; 5:436-49. [PMID: 25691969 PMCID: PMC4314274 DOI: 10.1002/ece3.1359] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 11/09/2014] [Accepted: 11/17/2014] [Indexed: 11/11/2022] Open
Abstract
Wildlife pathogens can alter host fitness. Low pathogenic avian influenza virus (LPAIV) infection is thought to have negligible impacts on wild birds; however, effects of infection in free-living birds are largely unstudied. We investigated the extent to which LPAIV infection and shedding were associated with body condition and immune status in free-living mallards (Anas platyrhynchos), a partially migratory key LPAIV host species. We sampled mallards throughout the species' annual autumn LPAIV infection peak, and we classified individuals according to age, sex, and migratory strategy (based on stable hydrogen isotope analysis) when analyzing data on body mass and five indices of immune status. Body mass was similar for LPAIV-infected and noninfected birds. The degree of virus shedding from the cloaca and oropharynx was not associated with body mass. LPAIV infection and shedding were not associated with natural antibody (NAbs) and complement titers (first lines of defense against infections), concentrations of the acute phase protein haptoglobin (Hp), ratios of heterophils to lymphocytes (H:L ratio), and avian influenza virus (AIV)-specific antibody concentrations. NAbs titers were higher in LPAIV-infected males and local (i.e., short distance) migrants than in infected females and distant (i.e., long distance) migrants. Hp concentrations were higher in LPAIV-infected juveniles and females compared to infected adults and males. NAbs, complement, and Hp levels were lower in LPAIV-infected mallards in early autumn. Our study demonstrates weak associations between infection with and shedding of LPAIV and the body condition and immune status of free-living mallards. These results may support the role of mallards as asymptomatic carriers of LPAIV and raise questions about possible coevolution between virus and host.
Collapse
Affiliation(s)
- Jacintha G B van Dijk
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW) Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus MC PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Marcel Klaassen
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW) Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands ; Centre for Integrative Ecology, School of Life and Environmental Sciences, Deakin University Locked Bag 20000, Geelong, Victoria, 3220, Australia
| | - Kevin D Matson
- Animal Ecology Group, Centre for Ecological and Evolutionary Studies, University of Groningen PO Box 11103, 9700 CC, Groningen, The Netherlands ; Resource Ecology Group, Wageningen University, Droevendaalsesteeg 3a 6708, PB Wageningen, The Netherlands
| |
Collapse
|
26
|
Kreijtz JHCM, Wiersma LCM, De Gruyter HLM, Vogelzang-van Trierum SE, van Amerongen G, Stittelaar KJ, Fouchier RAM, Osterhaus ADME, Sutter G, Rimmelzwaan GF. A single immunization with modified vaccinia virus Ankara-based influenza virus H7 vaccine affords protection in the influenza A(H7N9) pneumonia ferret model. J Infect Dis 2014; 211:791-800. [PMID: 25246535 DOI: 10.1093/infdis/jiu528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Since the first reports in early 2013, >440 human cases of infection with avian influenza A(H7N9) have been reported including 122 fatalities. After the isolation of the first A(H7N9) viruses, the nucleotide sequences became publically available. Based on the coding sequence of the influenza virus A/Shanghai/2/2013 hemagglutinin gene, a codon-optimized gene was synthesized and cloned into a recombinant modified vaccinia virus Ankara (MVA). This MVA-H7-Sh2 viral vector was used to immunize ferrets and proved to be immunogenic, even after a single immunization. Subsequently, ferrets were challenged with influenza virus A/Anhui/1/2013 via the intratracheal route. Unprotected animals that were mock vaccinated or received empty vector developed interstitial pneumonia characterized by a marked alveolitis, accompanied by loss of appetite, weight loss, and heavy breathing. In contrast, animals vaccinated with MVA-H7-Sh2 were protected from severe disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ron A M Fouchier
- Department of Viroscience, Erasmus Medical Center Viroclinics Biosciences, Rotterdam, the Netherlands Institute for Infectious Diseases and Zoonoses, LMU University of Munich German Center for Infection Research, Braunschweig, Germany
| | | | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, LMU University of Munich German Center for Infection Research, Braunschweig, Germany
| | | |
Collapse
|
27
|
Li C, Li C, Zhang AJX, To KKW, Lee ACY, Zhu H, Wu HWL, Chan JFW, Chen H, Hung IFN, Li L, Yuen KY. Avian influenza A H7N9 virus induces severe pneumonia in mice without prior adaptation and responds to a combination of zanamivir and COX-2 inhibitor. PLoS One 2014; 9:e107966. [PMID: 25232731 PMCID: PMC4169509 DOI: 10.1371/journal.pone.0107966] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/16/2014] [Indexed: 12/26/2022] Open
Abstract
Background Human infection caused by the avian influenza A H7N9 virus has a case-fatality rate of over 30%. Systematic study of the pathogenesis of avian H7N9 isolate and effective therapeutic strategies are needed. Methods BALB/c mice were inoculated intranasally with an H7N9 virus isolated from a chicken in a wet market epidemiologically linked to a fatal human case, (A/chicken/Zhejiang/DTID-ZJU01/2013 [CK1]), and with an H7N9 virus isolated from a human (A/Anhui/01/2013 [AH1]). The pulmonary viral loads, cytokine/chemokine profiles and histopathological changes of the infected mice were compared. The therapeutic efficacy of a non-steroidal anti-inflammatory drug (NSAID), celecoxib, was assessed. Results Without prior adaptation, intranasal inoculation of 106 plaque forming units (PFUs) of CK1 caused a mortality rate of 82% (14/17) in mice. Viral nucleoprotein and RNA expression were limited to the respiratory system and no viral RNA could be detected from brain, liver and kidney tissues. CK1 caused heavy alveolar inflammatory exudation and pulmonary hemorrhage, associated with high pulmonary levels of proinflammatory cytokines. In the mouse lung cell line LA-4, CK1 also induced high levels of interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2) mRNA. Administration of the antiviral zanamivir did not significantly improve survival in mice infected with CK1, but co-administration of the non-steroidal anti-inflammatory drug (NSAID) celecoxib in combination with zanamivir improved survival and lung pathology. Conclusions Our findings suggested that H7N9 viruses isolated from chicken without preceding trans-species adaptation can cause lethal mammalian pulmonary infection. The severe proinflammatory responses might be a factor contributing to the mortality. Treatment with combination of antiviral and NSAID could ameliorate pulmonary inflammation and may improve survival.
Collapse
MESH Headings
- Adaptation, Physiological/immunology
- Animals
- Antiviral Agents/pharmacology
- Antiviral Agents/therapeutic use
- Cell Line
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Cyclooxygenase 2 Inhibitors/therapeutic use
- Cytokines/metabolism
- Drug Evaluation, Preclinical
- Drug Synergism
- Drug Therapy, Combination
- Female
- Humans
- Influenza A Virus, H7N9 Subtype/drug effects
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/physiology
- Lung/pathology
- Lung/virology
- Mice
- Mice, Inbred BALB C
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/immunology
- Pneumonia, Viral/virology
- Virus Replication
- Zanamivir/pharmacology
- Zanamivir/therapeutic use
Collapse
Affiliation(s)
- Can Li
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Chuangen Li
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Anna J. X. Zhang
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- * E-mail: (K-YY); (AJXZ)
| | - Kelvin K. W. To
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Andrew C. Y. Lee
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Houshun Zhu
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hazel W. L. Wu
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Jasper F. W. Chan
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Honglin Chen
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Zhejiang University, Hangzhou, China
| | - Ivan F. N. Hung
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Zhejiang University, Hangzhou, China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Zhejiang University, Hangzhou, China
- * E-mail: (K-YY); (AJXZ)
| |
Collapse
|
28
|
Liu B, Havers F, Chen E, Yuan Z, Yuan H, Ou J, Shang M, Kang K, Liao K, Liu F, Li D, Ding H, Zhou L, Zhu W, Ding F, Zhang P, Wang X, Yao J, Xiang N, Zhou S, Liu X, Song Y, Su H, Wang R, Cai J, Cao Y, Wang X, Bai T, Wang J, Feng Z, Zhang Y, Widdowson MA, Li Q. Risk factors for influenza A(H7N9) disease--China, 2013. Clin Infect Dis 2014; 59:787-94. [PMID: 24928293 DOI: 10.1093/cid/ciu423] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The majority of human cases of novel avian influenza A(H7N9), which emerged in China in spring 2013, include reported exposure to poultry. However, specific host and exposure risk factors for disease are unknown, yet critical to design prevention measures. METHODS In April-June 2013, we conducted a case-control study in 8 Chinese provinces. Patients with laboratory-confirmed A(H7N9) (n = 89) were matched by age, sex, and neighborhood to controls (n = 339). Subjects completed a questionnaire on medical history and potential exposures, including poultry markets and other poultry exposure. We used conditional logistic regression to calculate matched and adjusted odds ratios (ORs) for the association of A(H7N9) virus infection with potential risk factors. RESULTS Fifty-five percent of patients compared with 31% of controls reported any contact with poultry (matched OR [mOR], 7.8; 95% confidence interval [CI], 3.3-18.8). Sixty-seven percent of patients compared with 35% of controls visited a live poultry market (mOR, 5.4; CI, 3.0-9.7). Visiting live poultry markets increased risk of infection even after adjusting for poultry contact and other confounders (adjusted OR, 3.4; CI, 1.8-6.7). Backyard poultry were not associated with increased risk; 14% of cases did not report any poultry exposure or market visit. Obesity (mOR, 4.7; CI, 1.8-12.4), chronic obstructive pulmonary disease (mOR, 2.7; CI, 1.1-6.9), and immunosuppressive medications (mOR, 9.0; CI, 1.7-47.2) were associated with A(H7N9) disease. CONCLUSION Exposures to poultry in markets were associated with A(H7N9) virus infection, even without poultry contact. China should consider permanently closing live poultry markets or aggressively pursuing control measures to prevent spread of this emerging pathogen.
Collapse
Affiliation(s)
- Bo Liu
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Fiona Havers
- Epidemic Intelligence Service assigned to the Influenza Division, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Enfu Chen
- Influenza Division, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou
| | - Zhengan Yuan
- Shanghai Municipal Center for Disease Control and Prevention
| | - Hui Yuan
- Jiangxi Provincial Center for Disease Control and Prevention, Nanchang
| | - Jianming Ou
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou
| | - Mei Shang
- China Office, US Centers for Disease Control and Prevention, Beijing
| | - Kai Kang
- Henan Provincial Center for Disease Control and Prevention, Zhengzhou
| | - Kaiju Liao
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Fuqiang Liu
- Hunan Provincial Center for Disease Control and Prevention, Changsha
| | - Dan Li
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Hua Ding
- Zhejiang Hangzhou Center for Disease Control and Prevention, Hangzhou
| | - Lei Zhou
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Weiping Zhu
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai
| | - Fan Ding
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Peng Zhang
- Zhejiang Huzhou Center for Disease Control and Prevention, Hangzhou
| | - Xiaoye Wang
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Jianyi Yao
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Nijuan Xiang
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Suizan Zhou
- China Office, US Centers for Disease Control and Prevention, Beijing
| | - Xiaoqin Liu
- Jiangxi Provincial Center for Disease Control and Prevention, Nanchang
| | - Ying Song
- China Office, US Centers for Disease Control and Prevention, Beijing
| | - Hualin Su
- Shanghai Minhang District Center for Disease Control and Prevention
| | - Rui Wang
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Jian Cai
- Influenza Division, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou
| | - Yang Cao
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Xianjun Wang
- Shandong Provincial Center for Disease Control and Prevention, Jinan City
| | - Tian Bai
- Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing
| | - Jianjun Wang
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| | - Zijian Feng
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Yanping Zhang
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| | - Marc-Alain Widdowson
- Influenza Division, US Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Qun Li
- Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing
| |
Collapse
|
29
|
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
|
30
|
Arilahti V, Mäkelä SM, Tynell J, Julkunen I, Österlund P. Novel avian influenza A (H7N9) virus induces impaired interferon responses in human dendritic cells. PLoS One 2014; 9:e96350. [PMID: 24804732 PMCID: PMC4012951 DOI: 10.1371/journal.pone.0096350] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/07/2014] [Indexed: 01/01/2023] Open
Abstract
In March 2013 a new avian influenza A(H7N9) virus emerged in China and infected humans with a case fatality rate of over 30%. Like the highly pathogenic H5N1 virus, H7N9 virus is causing severe respiratory distress syndrome in most patients. Based on genetic analysis this avian influenza A virus shows to some extent adaptation to mammalian host. In the present study, we analyzed the activation of innate immune responses by this novel H7N9 influenza A virus and compared these responses to those induced by the avian H5N1 and seasonal H3N2 viruses in human monocyte-derived dendritic cells (moDCs). We observed that in H7N9 virus-infected cells, interferon (IFN) responses were weak although the virus replicated as well as the H5N1 and H3N2 viruses in moDCs. H7N9 virus-induced expression of pro-inflammatory cytokines remained at a significantly lower level as compared to H5N1 virus-induced “cytokine storm” seen in human moDCs. However, the H7N9 virus was extremely sensitive to the antiviral effects of IFN-α and IFN-β in pretreated cells. Our data indicates that different highly pathogenic avian viruses may show considerable differences in their ability to induce host antiviral responses in human primary cell models such as moDCs. The unexpected appearance of the novel H7N9 virus clearly emphasizes the importance of the global influenza surveillance system. It is, however, equally important to systematically characterize in normal human cells the replication capacity of the new viruses and their ability to induce and respond to natural antiviral substances such as IFNs.
Collapse
Affiliation(s)
- Veera Arilahti
- Virology Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Sanna M. Mäkelä
- Virology Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Janne Tynell
- Virology Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Ilkka Julkunen
- Virology Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Virology, University of Turku, Turku, Finland
| | - Pamela Österlund
- Virology Unit, National Institute for Health and Welfare, Helsinki, Finland
- * E-mail:
| |
Collapse
|
31
|
Abdelwhab EM, Veits J, Mettenleiter TC. Prevalence and control of H7 avian influenza viruses in birds and humans. Epidemiol Infect 2014; 142:896-920. [PMID: 24423384 PMCID: PMC9151109 DOI: 10.1017/s0950268813003324] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/21/2013] [Accepted: 12/04/2013] [Indexed: 01/20/2023] Open
Abstract
The H7 subtype HA gene has been found in combination with all nine NA subtype genes. Most exhibit low pathogenicity and only rarely high pathogenicity in poultry (and humans). During the past few years infections of poultry and humans with H7 subtypes have increased markedly. This review summarizes the emergence of avian influenza virus H7 subtypes in birds and humans, and the possibilities of its control in poultry. All H7Nx combinations were reported from wild birds, the natural reservoir of the virus. Geographically, the most prevalent subtype is H7N7, which is endemic in wild birds in Europe and was frequently reported in domestic poultry, whereas subtype H7N3 is mostly isolated from the Americas. In humans, mild to fatal infections were caused by subtypes H7N2, H7N3, H7N7 and H7N9. While infections of humans have been associated mostly with exposure to domestic poultry, infections of poultry have been linked to wild birds or live-bird markets. Generally, depopulation of infected poultry was the main control tool; however, inactivated vaccines were also used. In contrast to recent cases caused by subtype H7N9, human infections were usually self-limiting and rarely required antiviral medication. Close genetic and antigenic relatedness of H7 viruses of different origins may be helpful in development of universal vaccines and diagnostics for both animals and humans. Due to the wide spread of H7 viruses and their zoonotic importance more research is required to better understand the epidemiology, pathobiology and virulence determinants of these viruses and to develop improved control tools.
Collapse
Affiliation(s)
- E M Abdelwhab
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Molecular Biology, Greifswald - Insel Riems, Germany
| | - J Veits
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Molecular Biology, Greifswald - Insel Riems, Germany
| | - T C Mettenleiter
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Molecular Biology, Greifswald - Insel Riems, Germany
| |
Collapse
|
32
|
Siegers JY, Short KR, Leijten LME, de Graaf M, Spronken MIJ, Schrauwen EJA, Marshall N, Lowen AC, Gabriel G, Osterhaus ADME, Kuiken T, van Riel D. Novel avian-origin influenza A (H7N9) virus attachment to the respiratory tract of five animal models. J Virol 2014; 88:4595-9. [PMID: 24478425 PMCID: PMC3993775 DOI: 10.1128/jvi.03190-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/27/2014] [Indexed: 11/20/2022] Open
Abstract
We determined the pattern of attachment of the avian-origin H7N9 influenza viruses A/Anhui/1/2013 and A/Shanghai/1/2013 to the respiratory tract in ferrets, macaques, mice, pigs, and guinea pigs and compared it to that in humans. The H7N9 attachment pattern in macaques, mice, and to a lesser extent pigs and guinea pigs resembled that in humans more closely than the attachment pattern in ferrets. This information contributes to our knowledge of the different animal models for influenza.
Collapse
Affiliation(s)
- Jurre Y. Siegers
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Kirsty R. Short
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | - Miranda de Graaf
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Nicolle Marshall
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anice C. Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Gülsah Gabriel
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | | | - Thijs Kuiken
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
33
|
de Graaf M, Fouchier RAM. Role of receptor binding specificity in influenza A virus transmission and pathogenesis. EMBO J 2014; 33:823-41. [PMID: 24668228 DOI: 10.1002/embj.201387442] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The recent emergence of a novel avian A/H7N9 influenza virus in poultry and humans in China, as well as laboratory studies on adaptation and transmission of avian A/H5N1 influenza viruses, has shed new light on influenza virus adaptation to mammals. One of the biological traits required for animal influenza viruses to cross the species barrier that received considerable attention in animal model studies, in vitro assays, and structural analyses is receptor binding specificity. Sialylated glycans present on the apical surface of host cells can function as receptors for the influenza virus hemagglutinin (HA) protein. Avian and human influenza viruses typically have a different sialic acid (SA)-binding preference and only few amino acid changes in the HA protein can cause a switch from avian to human receptor specificity. Recent experiments using glycan arrays, virus histochemistry, animal models, and structural analyses of HA have added a wealth of knowledge on receptor binding specificity. Here, we review recent data on the interaction between influenza virus HA and SA receptors of the host, and the impact on virus host range, pathogenesis, and transmission. Remaining challenges and future research priorities are also discussed.
Collapse
Affiliation(s)
- Miranda de Graaf
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
34
|
Detection of nonhemagglutinating influenza a(h3) viruses by enzyme-linked immunosorbent assay in quantitative influenza virus culture. J Clin Microbiol 2014; 52:1672-7. [PMID: 24622097 DOI: 10.1128/jcm.03575-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
To assess the efficacy of novel antiviral drugs against influenza virus in clinical trials, it is necessary to quantify infectious virus titers in respiratory tract samples from patients. Typically, this is achieved by inoculating virus-susceptible cells with serial dilutions of clinical specimens and detecting the production of progeny virus by hemagglutination, since influenza viruses generally have the capacity to bind and agglutinate erythrocytes of various species through their hemagglutinin (HA). This readout method is no longer adequate, since an increasing number of currently circulating influenza A virus H3 subtype (A[H3]) viruses display a reduced capacity to agglutinate erythrocytes. Here, we report the magnitude of this problem by analyzing the frequency of HA-deficient A(H3) viruses detected in The Netherlands from 1999 to 2012. Furthermore, we report the development and validation of an alternative method for monitoring the production of progeny influenza virus in quantitative virus cultures, which is independent of the capacity to agglutinate erythrocytes. This method is based on the detection of viral nucleoprotein (NP) in virus culture plates by enzyme-linked immunosorbent assay (ELISA), and it produced results similar to those of the hemagglutination assay using strains with good HA activity, including A/Brisbane/059/07 (H1N1), A/Victoria/210/09 (H3N2), other seasonal A(H1N1), A(H1N1)pdm09, and the majority of A(H3) virus strains isolated in 2009. In contrast, many A(H3) viruses that have circulated since 2010 failed to display HA activity, and infectious virus titers were determined only by detecting NP. The virus culture ELISA described here will enable efficacy testing of new antiviral compounds in clinical trials during seasons in which nonhemagglutinating influenza A viruses circulate.
Collapse
|
35
|
van de Sandt CE, Kreijtz JHCM, de Mutsert G, Geelhoed-Mieras MM, Hillaire MLB, Vogelzang-van Trierum SE, Osterhaus ADME, Fouchier RAM, Rimmelzwaan GF. Human cytotoxic T lymphocytes directed to seasonal influenza A viruses cross-react with the newly emerging H7N9 virus. J Virol 2014; 88:1684-93. [PMID: 24257602 PMCID: PMC3911609 DOI: 10.1128/jvi.02843-13] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/12/2013] [Indexed: 01/05/2023] Open
Abstract
In February 2013, zoonotic transmission of a novel influenza A virus of the H7N9 subtype was reported in China. Although at present no sustained human-to-human transmission has been reported, a pandemic outbreak of this H7N9 virus is feared. Since neutralizing antibodies to the hemagglutinin (HA) globular head domain of the virus are virtually absent in the human population, there is interest in identifying other correlates of protection, such as cross-reactive CD8(+) T cells (cytotoxic T lymphocytes [CTLs]) elicited during seasonal influenza A virus infections. These virus-specific CD8(+) T cells are known to recognize conserved internal proteins of influenza A viruses predominantly, but it is unknown to what extent they cross-react with the newly emerging H7N9 virus. Here, we assessed the cross-reactivity of seasonal H3N2 and H1N1 and pandemic H1N1 influenza A virus-specific polyclonal CD8(+) T cells, obtained from HLA-typed study subjects, with the novel H7N9 virus. The cross-reactivity of CD8(+) T cells to H7N9 variants of known influenza A virus epitopes and H7N9 virus-infected cells was determined by their gamma interferon (IFN-γ) response and lytic activity. It was concluded that, apart from recognition of individual H7N9 variant epitopes, CD8(+) T cells to seasonal influenza viruses display considerable cross-reactivity with the novel H7N9 virus. The presence of these cross-reactive CD8(+) T cells may afford some protection against infection with the new virus.
Collapse
MESH Headings
- Adult
- Amino Acid Sequence
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Cells, Cultured
- China/epidemiology
- Cross Protection
- Cross Reactions
- Disease Outbreaks
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/chemistry
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/chemistry
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/chemistry
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/isolation & purification
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/virology
- Interferon-gamma/immunology
- Male
- Middle Aged
- Molecular Sequence Data
- Seasons
- Sequence Alignment
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/virology
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ron A. M. Fouchier
- Viroscience Laboratory, Erasmus MC, Rotterdam, The Netherlands
- ViroClinics Biosciences BV, Rotterdam, The Netherlands
| | - Guus F. Rimmelzwaan
- Viroscience Laboratory, Erasmus MC, Rotterdam, The Netherlands
- ViroClinics Biosciences BV, Rotterdam, The Netherlands
| |
Collapse
|
36
|
Gabbard JD, Dlugolenski D, Van Riel D, Marshall N, Galloway SE, Howerth EW, Campbell PJ, Jones C, Johnson S, Byrd-Leotis L, Steinhauer DA, Kuiken T, Tompkins SM, Tripp R, Lowen AC, Steel J. Novel H7N9 influenza virus shows low infectious dose, high growth rate, and efficient contact transmission in the guinea pig model. J Virol 2014; 88:1502-12. [PMID: 24227867 PMCID: PMC3911619 DOI: 10.1128/jvi.02959-13] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/06/2013] [Indexed: 12/31/2022] Open
Abstract
The zoonotic outbreak of H7N9 subtype avian influenza virus that occurred in eastern China in the spring of 2013 resulted in 135 confirmed human cases, 44 of which were lethal. Sequencing of the viral genome revealed a number of molecular signatures associated with virulence or transmission in mammals. We report here that, in the guinea pig model, a human isolate of novel H7N9 influenza virus, A/Anhui/1/2013 (An/13), is highly dissimilar to an H7N1 avian isolate and instead behaves similarly to a human seasonal strain in several respects. An/13 was found to have a low 50% infectious dose, grow to high titers in the upper respiratory tract, and transmit efficiently among cocaged guinea pigs. The pH of fusion of the hemagglutinin (HA) and the binding of virus to fixed guinea pig tissues were also examined. The An/13 HA displayed a relatively elevated pH of fusion characteristic of many avian strains, and An/13 resembled avian viruses in terms of attachment to tissues. One important difference was seen between An/13 and both the H3N2 human and the H7N1 avian viruses: when inoculated intranasally at a high dose, only the An/13 virus led to productive infection of the lower respiratory tract of guinea pigs. In sum, An/13 was found to retain fusion and attachment properties of an avian influenza virus but displayed robust growth and contact transmission in the guinea pig model atypical of avian strains and indicative of mammalian adaptation.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Female
- Guinea Pigs
- Humans
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza A Virus, H7N1 Subtype/genetics
- Influenza A Virus, H7N1 Subtype/physiology
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/growth & development
- Influenza A Virus, H7N9 Subtype/pathogenicity
- Influenza A Virus, H7N9 Subtype/physiology
- Influenza, Human/transmission
- Influenza, Human/virology
- Virulence
Collapse
Affiliation(s)
- Jon D. Gabbard
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Daniel Dlugolenski
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Debby Van Riel
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nicolle Marshall
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Summer E. Galloway
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Patricia J. Campbell
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Cheryl Jones
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Scott Johnson
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Lauren Byrd-Leotis
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David A. Steinhauer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Thijs Kuiken
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - S. Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Ralph Tripp
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Anice C. Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - John Steel
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
37
|
Wang C, Yu H, Horby PW, Cao B, Wu P, Yang S, Gao H, Li H, Tsang TK, Liao Q, Gao Z, Ip DKM, Jia H, Jiang H, Liu B, Ni MY, Dai X, Liu F, Van Kinh N, Liem NT, Hien TT, Li Y, Yang J, Wu JT, Zheng Y, Leung GM, Farrar JJ, Cowling BJ, Uyeki TM, Li L. Comparison of patients hospitalized with influenza A subtypes H7N9, H5N1, and 2009 pandemic H1N1. Clin Infect Dis 2014; 58:1095-103. [PMID: 24488975 PMCID: PMC3967826 DOI: 10.1093/cid/ciu053] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Hospitalization with H7N9 virus infection is associated with older age and chronic heart
disease, and patients have a longer duration of hospitalization than patients with H5N1 or
pH1N1. This suggests that host factors are an important contributor to H7N9 severity. Background. Influenza A(H7N9) viruses isolated from
humans show features suggesting partial adaptation to mammals. To provide insights into
the pathogenesis of H7N9 virus infection, we compared risk factors, clinical presentation,
and progression of patients hospitalized with H7N9, H5N1, and 2009 pandemic H1N1 (pH1N1)
virus infections. Methods. We compared individual-level data from
patients hospitalized with infection by H7N9 (n = 123), H5N1 (n = 119; 43
China, 76 Vietnam), and pH1N1 (n = 3486) viruses. We assessed risk factors for
hospitalization after adjustment for age- and sex-specific prevalence of risk factors in
the general Chinese population. Results. The median age of patients with H7N9 virus
infection was older than other patient groups (63 years; P < .001) and
a higher proportion was male (71%; P < .02). After adjustment
for age and sex, chronic heart disease was associated with an increased risk of
hospitalization with H7N9 (relative risk, 9.68; 95% confidence interval,
5.24–17.9). H7N9 patients had similar patterns of leukopenia, thrombocytopenia, and
elevated alanine aminotransferase, creatinine kinase, C-reactive protein, and lactate
dehydrogenase to those seen in H5N1 patients, which were all significantly different from
pH1N1 patients (P < .005). H7N9 patients had a longer duration of
hospitalization than either H5N1 or pH1N1 patients (P < .001), and the
median time from onset to death was 18 days for H7N9 (P = .002) vs
11 days for H5N1 and 15 days for pH1N1 (P = .154). Conclusions. The identification of known risk factors
for severe seasonal influenza and the more protracted clinical course compared with that
of H5N1 suggests that host factors are an important contributor to H7N9 severity.
Collapse
Affiliation(s)
- Chen Wang
- Institute of Respiratory Medicine, Beijing Hospital, National Health and Family Planning Commission
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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
|