201
|
Xu L, Bao L, Deng W, Zhu H, Li F, Chen T, Lv Q, Yuan J, Xu Y, Li Y, Yao Y, Gu S, Yu P, Chen H, Qin C. Rapid adaptation of avian H7N9 virus in pigs. Virology 2014; 452-453:231-6. [DOI: 10.1016/j.virol.2014.01.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/13/2013] [Accepted: 01/20/2014] [Indexed: 12/01/2022]
|
202
|
Pasricha G, Mukherjee S, Chakrabarti AK. Comprehensive sequence analysis of HA protein of H7 subtype avian influenza viruses: an emphasis on mutations in novel H7N9 viruses. Future Virol 2014. [DOI: 10.2217/fvl.13.132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: Aim: H7 avian influenza viruses pose a major public health threat raising concerns regarding their pandemic potential, especially after the recent outbreak in China of H7N9 subtype viruses. The objective was to gain insight into the geographical and host-wise distribution of H7 subtype viruses, and to understand molecular determinants responsible for their adaptation in humans. Materials & methods: In this study we carried out a global comprehensive analysis of 1749 HA sequences belonging to the H7 subtype available in the Global Initiative on Sharing All Influenza Data (GISAID) EpiFlu™ database. We also analyzed full-genome sequences of the 27 influenza strains belonging to the H7N9 subtype isolated recently from China. Results: Most of the H7 strains were from North America (749) followed by in Europe (659) and Asia (284). The majority of the sequences belonged to the H7N7 subtype (524) followed by H7N3 (440) and H7N2 (411), while 107 belonged to H7N9. Comparison of HA sequences of H7 viruses isolated from humans showed the presence of mutations and determinants that could have played a pivotal role in avian-to-human transmission and adaptability in humans. Mutational analysis of all the segments of the recent H7N9 viruses isolated from humans in China revealed that these viruses possessed several characteristic features of mammalian influenza viruses. Conclusion: H7 viruses, irrespective of being of high or low pathogenicity have a propensity to adapt to humans causing mild to severe infections. These viruses have signature mutations that have been associated with interspecies transmission and human adaptability, raising concerns regarding their pandemic potential.
Collapse
Affiliation(s)
- Gunisha Pasricha
- Microbial Containment Complex, National Institute of Virology, Sus Road, Pashan, Pune, 411021, India
| | - Sanjay Mukherjee
- Microbial Containment Complex, National Institute of Virology, Sus Road, Pashan, Pune, 411021, India
| | - Alok K Chakrabarti
- Microbial Containment Complex, National Institute of Virology, Sus Road, Pashan, Pune, 411021, India
| |
Collapse
|
203
|
Yamayoshi S, Yamada S, Fukuyama S, Murakami S, Zhao D, Uraki R, Watanabe T, Tomita Y, Macken C, Neumann G, Kawaoka Y. Virulence-affecting amino acid changes in the PA protein of H7N9 influenza A viruses. J Virol 2014; 88:3127-34. [PMID: 24371069 PMCID: PMC3957961 DOI: 10.1128/jvi.03155-13] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED Novel avian-origin influenza A(H7N9) viruses were first reported to infect humans in March 2013. To date, 143 human cases, including 45 deaths, have been recorded. By using sequence comparisons and phylogenetic and ancestral inference analyses, we identified several distinct amino acids in the A(H7N9) polymerase PA protein, some of which may be mammalian adapting. Mutant viruses possessing some of these amino acid changes, singly or in combination, were assessed for their polymerase activities and growth kinetics in mammalian and avian cells and for their virulence in mice. We identified several mutants that were slightly more virulent in mice than the wild-type A(H7N9) virus, A/Anhui/1/2013. These mutants also exhibited increased polymerase activity in human cells but not in avian cells. Our findings indicate that the PA protein of A(H7N9) viruses has several amino acid substitutions that are attenuating in mammals. IMPORTANCE Novel avian-origin influenza A(H7N9) viruses emerged in the spring of 2013. By using computational analyses of A(H7N9) viral sequences, we identified several amino acid changes in the polymerase PA protein, which we then assessed for their effects on viral replication in cultured cells and mice. We found that the PA proteins of A(H7N9) viruses possess several amino acid substitutions that cause attenuation in mammals.
Collapse
Affiliation(s)
- Seiya Yamayoshi
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shinya Yamada
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoshi Fukuyama
- ERATO Infection-Induced Host Responses Project, Japan Science and Technology Agency, Saitama, Japan
| | - Shin Murakami
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Dongming Zhao
- ERATO Infection-Induced Host Responses Project, Japan Science and Technology Agency, Saitama, Japan
| | - Ryuta Uraki
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tokiko Watanabe
- ERATO Infection-Induced Host Responses Project, Japan Science and Technology Agency, Saitama, Japan
| | - Yuriko Tomita
- ERATO Infection-Induced Host Responses Project, Japan Science and Technology Agency, Saitama, Japan
| | - Catherine Macken
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Gabriele Neumann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- ERATO Infection-Induced Host Responses Project, Japan Science and Technology Agency, Saitama, Japan
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| |
Collapse
|
204
|
Host adaptation and transmission of influenza A viruses in mammals. Emerg Microbes Infect 2014; 3:e9. [PMID: 26038511 PMCID: PMC3944123 DOI: 10.1038/emi.2014.9] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/11/2013] [Accepted: 12/13/2013] [Indexed: 12/17/2022]
Abstract
A wide range of influenza A viruses of pigs and birds have infected humans in the last decade, sometimes with severe clinical consequences. Each of these so-called zoonotic infections provides an opportunity for virus adaptation to the new host. Fortunately, most of these human infections do not yield viruses with the ability of sustained human-to-human transmission. However, animal influenza viruses have acquired the ability of sustained transmission between humans to cause pandemics on rare occasions in the past, and therefore, influenza virus zoonoses continue to represent threats to public health. Numerous recent studies have shed new light on the mechanisms of adaptation and transmission of avian and swine influenza A viruses in mammals. In particular, several studies provided insights into the genetic and phenotypic traits of influenza A viruses that may determine airborne transmission. Here, we summarize recent studies on molecular determinants of virulence and adaptation of animal influenza A virus and discuss the phenotypic traits associated with airborne transmission of newly emerging influenza A viruses. Increased understanding of the determinants and mechanisms of virulence and transmission may aid in assessing the risks posed by animal influenza viruses to human health, and preparedness for such risks.
Collapse
|
205
|
TMPRSS2 is a host factor that is essential for pneumotropism and pathogenicity of H7N9 influenza A virus in mice. J Virol 2014; 88:4744-51. [PMID: 24522916 DOI: 10.1128/jvi.03799-13] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED Cleavage of the hemagglutinin (HA) by host proteases is essential for the infectivity of influenza viruses. Here, we analyzed the role of the serine protease TMPRSS2, which activates HA in the human respiratory tract, in pathogenesis in a mouse model. Replication of the human H7N9 isolate A/Anhui/1/13 and of human H1N1 and H3N2 viruses was compared in TMPRSS2 knockout (TMPRSS2(-/-)) and wild-type (WT) mice. Knockout of TMPRSS2 expression inhibited H7N9 influenza virus replication in explants of murine tracheas, bronchi, and lungs. H1N1 virus replication was also strongly suppressed in airway explants of TMPRSS2(-/-) mice, while H3N2 virus replication was only marginally affected. H7N9 and H1N1 viruses were apathogenic in TMPRSS2(-/-) mice, whereas WT mice developed severe disease with mortality rates of 100% and 20%, respectively. In contrast, all H3N2 infected TMPRSS2(-/-) and WT mice succumbed to lethal infection. Cleavage analysis showed that H7 and H1 are efficiently activated by TMPRSS2, whereas H3 is less susceptible to the protease. Our data demonstrate that TMPRSS2 is a host factor that is essential for pneumotropism and pathogenicity of H7N9 and H1N1 influenza virus in mice. In contrast, replication of H3N2 virus appears to depend on another, not yet identified protease, supporting the concept that human influenza viruses differ in protease specificity. IMPORTANCE Cleavage of the hemagglutinin (HA) by host proteases is essential for the infectivity of influenza virus, but little is known about its relevance for pathogenesis in mammals. Here, we show that knockout mice that do not express the HA-activating protease TMPRSS2 are resistant to pulmonary disease with lethal outcome when infected with influenza A viruses of subtypes H7N9 and H1N1, whereas they are not protected from lethal H3N2 virus infection. These findings demonstrate that human influenza viruses differ in protease specificity, and that expression of the appropriate protease in respiratory tissues is essential for pneumotropism and pathogenicity. Our observations also demonstrate that HA-activating proteases and in particular TMPRSS2 are promising targets for influenza therapy.
Collapse
|
206
|
Transcriptomic characterization of the novel avian-origin influenza A (H7N9) virus: specific host response and responses intermediate between avian (H5N1 and H7N7) and human (H3N2) viruses and implications for treatment options. mBio 2014; 5:e01102-13. [PMID: 24496798 PMCID: PMC3950506 DOI: 10.1128/mbio.01102-13] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED A novel avian-origin H7N9 influenza A virus (IAV) emerged in China in 2013, causing mild to lethal human respiratory infections. H7N9 originated with multiple reassortment events between avian viruses and carries genetic markers of human adaptation. Determining whether H7N9 induces a host response closer to that with human or avian IAV is important in order to better characterize this emerging virus. Here we compared the human lung epithelial cell response to infection with A/Anhui/01/13 (H7N9) or highly pathogenic avian-origin H5N1, H7N7, or human seasonal H3N2 IAV. The transcriptomic response to H7N9 was highly specific to this strain but was more similar to the response to human H3N2 than to that to other avian IAVs. H7N9 and H3N2 both elicited responses related to eicosanoid signaling and chromatin modification, whereas H7N9 specifically induced genes regulating the cell cycle and transcription. Among avian IAVs, the response to H7N9 was closest to that elicited by H5N1 virus. Host responses common to H7N9 and the other avian viruses included the lack of induction of the antigen presentation pathway and reduced proinflammatory cytokine induction compared to that with H3N2. Repression of these responses could have an important impact on the immunogenicity and virulence of H7N9 in humans. Finally, using a genome-based drug repurposing approach, we identified several drugs predicted to regulate the host response to H7N9 that may act as potential antivirals, including several kinase inhibitors, as well as FDA-approved drugs, such as troglitazone and minocycline. Importantly, we validated that minocycline inhibited H7N9 replication in vitro, suggesting that our computational approach holds promise for identifying novel antivirals. IMPORTANCE Whether H7N9 will be the next pandemic influenza virus or will persist and sporadically infect humans from its avian reservoir, similar to H5N1, is not known yet. High-throughput profiling of the host response to infection allows rapid characterization of virus-host interactions and generates many hypotheses that will accelerate understanding and responsiveness to this potential threat. We show that the cellular response to H7N9 virus is closer to that induced by H3N2 than to that induced by H5N1, reflecting the potential of this new virus for adaptation to humans. Importantly, dissecting the host response to H7N9 may guide host-directed antiviral development.
Collapse
|
207
|
Qiu C, Yuan S, Tian D, Yang Y, Zhang A, Chen Q, Wan Y, Song Z, He J, Li L, Sun J, Zhou M, Qiu C, Zhang Z, Lu S, Zhang X, Hu Y, Xu J. Epidemiologic report and serologic findings for household contacts of three cases of influenza A (H7N9) virus infection. J Clin Virol 2014; 59:129-31. [PMID: 24388209 PMCID: PMC7106538 DOI: 10.1016/j.jcv.2013.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 11/23/2013] [Accepted: 12/04/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE We conducted epidemiologic investigations and serologic assays on household contacts that were extensively exposed to three influenza A (H7N9) virus infected case-patients before infection-control practices were implemented. STUDY DESIGN Data on the early clinical course of each patient and the exposure history for each patient's household contacts were obtained by interviewing household members and by reviewing medical records. Viral RNA in patient samples was tested using real-time reverse transcriptase polymerase chain reaction assay. Antibodies against H7N9 virus in serum samples were tested using hemagglutination inhibition and pseudovirus based neutralization assays. RESULTS All household contacts were extensively exposed to the case-patients without the use of measures to protect against infection. Viral RNA was detected in the specimens from case-patients for approximately 7-11 days after confirmation of infection. However, the results of the analyses of serum specimens taken from the household contacts 15-26 days post exposure revealed no evidence of transmission of H7N9 virus from the case-patients to the contacts. CONCLUSION Despite ample unprotected exposures to case-patients during the virus shedding period, household members in this report were not infected by the H7N9 virus.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Viral/blood
- Contact Tracing
- Family Characteristics
- Female
- Hemagglutination Inhibition Tests
- Humans
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/isolation & purification
- Influenza, Human/epidemiology
- Influenza, Human/transmission
- Influenza, Human/virology
- Male
- Middle Aged
- Neutralization Tests
- RNA, Viral/blood
- RNA, Viral/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Chao Qiu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Songhua Yuan
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Di Tian
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Yang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anli Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingguo Chen
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanmin Wan
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhigang Song
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing He
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liangzhu Li
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Sun
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingzhe Zhou
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenli Qiu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyong Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuihua Lu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China; State Key Laboratory for Infectious Disease Prevention and Control, China CDC, Beijing, China.
| | - Yunwen Hu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jianqing Xu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China; State Key Laboratory for Infectious Disease Prevention and Control, China CDC, Beijing, China.
| |
Collapse
|
208
|
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: 8.1] [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
|
209
|
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: 3.9] [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
|
210
|
Hsu Y, Ma HH, Lico LS, Jan JT, Fukase K, Uchinashi Y, Zulueta MML, Hung SC. One-pot synthesis of N-acetyl- and N-glycolylneuraminic acid capped trisaccharides and evaluation of their influenza A(H1 N1) inhibition. Angew Chem Int Ed Engl 2014; 53:2413-6. [PMID: 24482157 DOI: 10.1002/anie.201309646] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/25/2013] [Indexed: 12/23/2022]
Abstract
Human lung epithelial cells natively offer terminal N-acetylneuraminic acid (Neu5Ac) α(2→6)-linked to galactose (Gal) as binding sites for influenza virus hemagglutinin. N-Glycolylneuraminic acid (Neu5Gc) in place of Neu5Ac is known to affect hemagglutinin binding in other species. Not normally generated by humans, Neu5Gc may find its way to human cells from dietary sources. To compare their influence in influenza virus infection, six trisaccharides with Neu5Ac or Neu5Gc α(2→6) linked to Gal and with different reducing end sugar units were prepared using one-pot assembly and divergent transformation. The sugar assembly made use of an N-phthaloyl-protected sialyl imidate for chemoselective activation and α-stereoselective coupling with a thiogalactoside. Assessment of cytopathic effect showed that the Neu5Gc-capped trisaccharides inhibited the viral infection better than their Neu5Ac counterparts.
Collapse
Affiliation(s)
- Yun Hsu
- Genomics Research Center, Academia Sinica, No. 128 Academia Road, Section 2, Taipei 115 (Taiwan); Department of Chemistry, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300 (Taiwan)
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Hsu Y, Ma HH, Lico LS, Jan JT, Fukase K, Uchinashi Y, Zulueta MML, Hung SC. One-Pot Synthesis ofN-Acetyl- andN-Glycolylneuraminic Acid Capped Trisaccharides and Evaluation of Their Influenza A(H1 N1) Inhibition. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201309646] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
212
|
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: 8.1] [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
|
213
|
Wu YL, Shen LW, Ding YP, Tanaka Y, Zhang W. Preliminary success in the characterization and management of a sudden breakout of a novel H7N9 influenza A virus. Int J Biol Sci 2014; 10:109-18. [PMID: 24520209 PMCID: PMC3920865 DOI: 10.7150/ijbs.8198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/05/2013] [Indexed: 12/21/2022] Open
Abstract
Influenza has always been one of the major threats to human health. The Spanish influenza in 1918, the pandemic influenza A/H1N1 in 2009, and the avian influenza A/H5N1 have brought about great disasters or losses to mankind. More recently, a novel avian influenza A/H7N9 broke out in China and until December 2, 2013, it had caused 139 cases of infection, including 45 deaths. Its risk and pandemic potential attract worldwide attention. In this article, we summarize epidemiology, virology characteristics, clinical symptoms, diagnosis methods, clinical treatment and preventive measures about the avian influenza A/H7N9 virus infection to provide a reference for a possible next wave of flu outbreak.
Collapse
Affiliation(s)
- Yan-Ling Wu
- 1. Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, 630 Xincheng Road, Hangzhou, 310051, PR China
| | - Li-Wen Shen
- 2. Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, PR China
| | - Yan-Ping Ding
- 1. Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, 630 Xincheng Road, Hangzhou, 310051, PR China
- 2. Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, PR China
| | - Yoshimasa Tanaka
- 3. Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Wen Zhang
- 2. Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, PR China
| |
Collapse
|
214
|
Preexisting CD8+ T-cell immunity to the H7N9 influenza A virus varies across ethnicities. Proc Natl Acad Sci U S A 2014; 111:1049-54. [PMID: 24395804 DOI: 10.1073/pnas.1322229111] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The absence of preexisting neutralizing antibodies specific for the novel A (H7N9) influenza virus indicates a lack of prior human exposure. As influenza A virus-specific CD8(+) T lymphocytes (CTLs) can be broadly cross-reactive, we tested whether immunogenic peptides derived from H7N9 might be recognized by memory CTLs established following infection with other influenza strains. Probing across multiple ethnicities, we identified 32 conserved epitopes derived from the nucleoprotein (NP) and matrix-1 (M1) proteins. These NP and M1 peptides are presented by HLAs prevalent in 16-57% of individuals. Remarkably, some HLA alleles (A*0201, A*0301, B*5701, B*1801, and B*0801) elicit robust CTL responses against any human influenza A virus, including H7N9, whereas ethnicities where HLA-A*0101, A*6801, B*1501, and A*2402 are prominent, show limited CTL response profiles. By this criterion, some groups, especially the Alaskan and Australian Indigenous peoples, would be particularly vulnerable to H7N9 infection. This dissection of CTL-mediated immunity to H7N9 thus suggests strategies for both vaccine delivery and development.
Collapse
|
215
|
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: 1.2] [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
|
216
|
Li C, Chen H. Enhancement of influenza virus transmission by gene reassortment. Curr Top Microbiol Immunol 2014; 385:185-204. [PMID: 25048543 DOI: 10.1007/82_2014_389] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Influenza A virus is characterized by a genome composed of eight single-stranded, negative sense RNA segments, which allow for reassortment between different strains when they co-infect the same host cell. Reassortment is an important driving force for the evolution of influenza viruses. The ability of reassortment allows influenza virus to endlessly reinvent itself and pose a constant threat to the health of humans and other animals. Of the four human influenza pandemics since the beginning of the last century, three of them were caused by reassortant viruses bearing genes of avian, human or swine influenza virus origin. In the past decade, great efforts have been made to understand the transmissibility of influenza viruses. The use of reverse genetics technology has made it substantially easier to generate reassortant viruses and evaluate the contribution of individual virus gene on virus transmissibility in animal models such as ferrets and guinea pigs. H5, H7, and H9 avian influenza viruses represent the top three subtypes that are candidates to cause the next human influenza pandemic. Many studies have been conducted to determine whether the transmission of these avian influenza viruses could be enhanced by acquisition of gene segments from human influenza viruses. Moreover, the 2009 pdmH1N1 viruses and the triple reassortant swine influenza viruses were extensively studied to identify the gene segments that contribute to their transmissibility. These studies have greatly deepened our understanding of the transmissibility of reassortant influenza viruses, which, in turn, has improved our ability to be prepared for reassortant influenza virus with enhanced transmissibility and pandemic potential.
Collapse
Affiliation(s)
- Chengjun Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 427 Maduan Street, Harbin, 150001, Nangang, China,
| | | |
Collapse
|
217
|
Influenza A☆. REFERENCE MODULE IN BIOMEDICAL SCIENCES 2014. [PMCID: PMC7157457 DOI: 10.1016/b978-0-12-801238-3.05048-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The influenza viruses, which contain single-stranded RNA, are classified into three types, A, B, and C and belong to the Orthomyxoviridae family of viruses. Types A and B cause annual epidemics and often pandemics of influenza illness, while type C is a less common disease with fewer symptoms.
Collapse
|
218
|
Herfst S, Imai M, Kawaoka Y, Fouchier RAM. Avian influenza virus transmission to mammals. Curr Top Microbiol Immunol 2014; 385:137-55. [PMID: 25048542 DOI: 10.1007/82_2014_387] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Influenza A viruses cause yearly epidemics and occasional pandemics. In addition, zoonotic influenza A viruses sporadically infect humans and may cause severe respiratory disease and fatalities. Fortunately, most of these viruses do not have the ability to be efficiently spread among humans via aerosols or respiratory droplets (airborne transmission) and to subsequently cause a pandemic. However, adaptation of these zoonotic viruses to humans by mutation or reassortment with human influenza A viruses may result in airborne transmissible viruses with pandemic potential. Although our knowledge of factors that affect mammalian adaptation and transmissibility of influenza viruses is still limited, we are beginning to understand some of the biological traits that drive airborne transmission of influenza viruses among mammals. Increased understanding of the determinants and mechanisms of airborne transmission may aid in assessing the risks posed by avian influenza viruses to human health, and preparedness for such risks. This chapter summarizes recent discoveries on the genetic and phenotypic traits required for avian influenza viruses to become airborne transmissible between mammals.
Collapse
Affiliation(s)
- S Herfst
- Department of Viroscience, Postgraduate School Molecular Medicine, Erasmus Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
219
|
Xu R, de Vries RP, Zhu X, Nycholat CM, McBride R, Yu W, Paulson JC, Wilson IA. Preferential recognition of avian-like receptors in human influenza A H7N9 viruses. Science 2013; 342:1230-5. [PMID: 24311689 DOI: 10.1126/science.1243761] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The 2013 outbreak of avian-origin H7N9 influenza in eastern China has raised concerns about its ability to transmit in the human population. The hemagglutinin glycoprotein of most human H7N9 viruses carries Leu(226), a residue linked to adaptation of H2N2 and H3N2 pandemic viruses to human receptors. However, glycan array analysis of the H7 hemagglutinin reveals negligible binding to humanlike α2-6-linked receptors and strong preference for a subset of avian-like α2-3-linked glycans recognized by all avian H7 viruses. Crystal structures of H7N9 hemagglutinin and six hemagglutinin-glycan complexes have elucidated the structural basis for preferential recognition of avian-like receptors. These findings suggest that the current human H7N9 viruses are poorly adapted for efficient human-to-human transmission.
Collapse
Affiliation(s)
- Rui Xu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
220
|
Liu Q, Ma J, Strayer DR, Mitchell WM, Carter WA, Ma W, Richt JA. Emergence of a novel drug resistant H7N9 influenza virus: evidence based clinical potential of a natural IFN-α for infection control and treatment. Expert Rev Anti Infect Ther 2013; 12:165-9. [PMID: 24350808 DOI: 10.1586/14787210.2014.870885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The novel avian H7N9 influenza virus has caused more than 130 human infections with 43 deaths (as of September, 2013) in China. Because of the lack of existing immunity against H7 subtype influenza viruses in the human population and the absence of a licensed commercial vaccine, antiviral drugs are critical tools for the treatment of infection with this novel H7N9. Both M2-ion channel blockers and neuraminidase inhibitors are used as antiviral drugs for influenza infections of humans. The emerging H7N9 viruses are resistant to the M2-ion channel blockers because of a S31N mutation in the M2 protein; additionally, some H7N9 isolates have gained neuraminidase R292K substitution resulting in broad resistance to neuraminidase inhibitors. In this study we report that Alferon N can inhibit wild type and 292K H7N9 viruses replication in vitro. Since Alferon N is approved for clinical use, this would allow a rapid regulatory approval process for this drug under pandemic threat.
Collapse
Affiliation(s)
- Qinfang Liu
- Center of Excellence for Emerging and Zoonotic Animal Diseases (CEEZAD), Kansas State University, Manhattan, KS 66506-5601, USA
| | | | | | | | | | | | | |
Collapse
|
221
|
|
222
|
Dortmans JCFM, Dekkers J, Wickramasinghe INA, Verheije MH, Rottier PJM, van Kuppeveld FJM, de Vries E, de Haan CAM. Adaptation of novel H7N9 influenza A virus to human receptors. Sci Rep 2013; 3:3058. [PMID: 24162312 PMCID: PMC3808826 DOI: 10.1038/srep03058] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/11/2013] [Indexed: 12/22/2022] Open
Abstract
The emergence of the novel H7N9 influenza A virus (IAV) has caused global concerns about the ability of this virus to spread between humans. Analysis of the receptor-binding properties of this virus using a recombinant protein approach in combination with fetuin-binding, glycan array and human tissue-binding assays demonstrates increased binding of H7 to both α2-6 and α2-8 sialosides as well as reduced binding to α2-3-linked SIAs compared to a closely related avian H7N9 virus from 2008. These differences could be attributed to substitutions Q226L and G186V. Analysis of the enzymatic activity of the neuraminidase N9 protein indicated a reduced sialidase activity, consistent with the reduced binding of H7 to α2-3 sialosides. However, the novel H7N9 virus still preferred binding to α2-3- over α2-6-linked SIAs and was not able to efficiently bind to epithelial cells of human trachea in contrast to seasonal IAV, consistent with its limited human-to-human transmission.
Collapse
Affiliation(s)
- J C F M Dortmans
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
223
|
Lam TTY, Wang J, Shen Y, Zhou B, Duan L, Cheung CL, Ma C, Lycett SJ, Leung CYH, Chen X, Li L, Hong W, Chai Y, Zhou L, Liang H, Ou Z, Liu Y, Farooqui A, Kelvin DJ, Poon LLM, Smith DK, Pybus OG, Leung GM, Shu Y, Webster RG, Webby RJ, Peiris JSM, Rambaut A, Zhu H, Guan Y. The genesis and source of the H7N9 influenza viruses causing human infections in China. Nature 2013; 502:241-4. [PMID: 23965623 PMCID: PMC3801098 DOI: 10.1038/nature12515] [Citation(s) in RCA: 376] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/01/2013] [Indexed: 02/05/2023]
Abstract
A novel H7N9 influenza A virus first detected in March 2013 has since caused more than 130 human infections in China, resulting in 40 deaths. Preliminary analyses suggest that the virus is a reassortant of H7, N9 and H9N2 avian influenza viruses, and carries some amino acids associated with mammalian receptor binding, raising concerns of a new pandemic. However, neither the source populations of the H7N9 outbreak lineage nor the conditions for its genesis are fully known. Using a combination of active surveillance, screening of virus archives, and evolutionary analyses, here we show that H7 viruses probably transferred from domestic duck to chicken populations in China on at least two independent occasions. We show that the H7 viruses subsequently reassorted with enzootic H9N2 viruses to generate the H7N9 outbreak lineage, and a related previously unrecognized H7N7 lineage. The H7N9 outbreak lineage has spread over a large geographic region and is prevalent in chickens at live poultry markets, which are thought to be the immediate source of human infections. Whether the H7N9 outbreak lineage has, or will, become enzootic in China and neighbouring regions requires further investigation. The discovery here of a related H7N7 influenza virus in chickens that has the ability to infect mammals experimentally, suggests that H7 viruses may pose threats beyond the current outbreak. The continuing prevalence of H7 viruses in poultry could lead to the generation of highly pathogenic variants and further sporadic human infections, with a continued risk of the virus acquiring human-to-human transmissibility.
Collapse
Affiliation(s)
- Tommy Tsan-Yuk Lam
- Joint Influenza Research Centre (SUMC/HKU), Shantou University Medical College, Shantou 515041, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
The novel human influenza A(H7N9) virus is naturally adapted to efficient growth in human lung tissue. mBio 2013; 4:e00601-13. [PMID: 24105764 PMCID: PMC3791893 DOI: 10.1128/mbio.00601-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A novel influenza A virus (IAV) of the H7N9 subtype has been isolated from severely diseased patients with pneumonia and acute respiratory distress syndrome and, apparently, from healthy poultry in March 2013 in Eastern China. We evaluated replication, tropism, and cytokine induction of the A/Anhui/1/2013 (H7N9) virus isolated from a fatal human infection and two low-pathogenic avian H7 subtype viruses in a human lung organ culture system mimicking infection of the lower respiratory tract. The A(H7N9) patient isolate replicated similarly well as a seasonal IAV in explanted human lung tissue, whereas avian H7 subtype viruses propagated poorly. Interestingly, the avian H7 strains provoked a strong antiviral type I interferon (IFN-I) response, whereas the A(H7N9) virus induced only low IFN levels. Nevertheless, all viruses analyzed were detected predominantly in type II pneumocytes, indicating that the A(H7N9) virus does not differ in its cellular tropism from other avian or human influenza viruses. Tissue culture-based studies suggested that the low induction of the IFN-β promoter correlated with an efficient suppression by the viral NS1 protein. These findings demonstrate that the zoonotic A(H7N9) virus is unusually well adapted to efficient propagation in human alveolar tissue, which most likely contributes to the severity of lower respiratory tract disease seen in many patients. Humans are usually not infected by avian influenza A viruses (IAV), but this large group of viruses contributes to the emergence of human pandemic strains. Transmission of virulent avian IAV to humans is therefore an alarming event that requires assessment of the biology as well as pathogenic and pandemic potentials of the viruses in clinically relevant models. Here, we demonstrate that an early virus isolate from the recent A(H7N9) outbreak in Eastern China replicated as efficiently as human-adapted IAV in explanted human lung tissue, whereas avian H7 subtype viruses were unable to propagate. Robust replication of the H7N9 strain correlated with a low induction of antiviral beta interferon (IFN-β), and cell-based studies indicated that this is due to efficient suppression of the IFN response by the viral NS1 protein. Thus, explanted human lung tissue appears to be a useful experimental model to explore the determinants facilitating cross-species transmission of the H7N9 virus to humans.
Collapse
|
225
|
Chowell G, Simonsen L, Towers S, Miller MA, Viboud C. Transmission potential of influenza A/H7N9, February to May 2013, China. BMC Med 2013; 11:214. [PMID: 24083506 PMCID: PMC3851127 DOI: 10.1186/1741-7015-11-214] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/30/2013] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND On 31 March 2013, the first human infections with the novel influenza A/H7N9 virus were reported in Eastern China. The outbreak expanded rapidly in geographic scope and size, with a total of 132 laboratory-confirmed cases reported by 3 June 2013, in 10 Chinese provinces and Taiwan. The incidence of A/H7N9 cases has stalled in recent weeks, presumably as a consequence of live bird market closures in the most heavily affected areas. Here we compare the transmission potential of influenza A/H7N9 with that of other emerging pathogens and evaluate the impact of intervention measures in an effort to guide pandemic preparedness. METHODS We used a Bayesian approach combined with a SEIR (Susceptible-Exposed-Infectious-Removed) transmission model fitted to daily case data to assess the reproduction number (R) of A/H7N9 by province and to evaluate the impact of live bird market closures in April and May 2013. Simulation studies helped quantify the performance of our approach in the context of an emerging pathogen, where human-to-human transmission is limited and most cases arise from spillover events. We also used alternative approaches to estimate R based on individual-level information on prior exposure and compared the transmission potential of influenza A/H7N9 with that of other recent zoonoses. RESULTS Estimates of R for the A/H7N9 outbreak were below the epidemic threshold required for sustained human-to-human transmission and remained near 0.1 throughout the study period, with broad 95% credible intervals by the Bayesian method (0.01 to 0.49). The Bayesian estimation approach was dominated by the prior distribution, however, due to relatively little information contained in the case data. We observe a statistically significant deceleration in growth rate after 6 April 2013, which is consistent with a reduction in A/H7N9 transmission associated with the preemptive closure of live bird markets. Although confidence intervals are broad, the estimated transmission potential of A/H7N9 appears lower than that of recent zoonotic threats, including avian influenza A/H5N1, swine influenza H3N2sw and Nipah virus. CONCLUSION Although uncertainty remains high in R estimates for H7N9 due to limited epidemiological information, all available evidence points to a low transmission potential. Continued monitoring of the transmission potential of A/H7N9 is critical in the coming months as intervention measures may be relaxed and seasonal factors could promote disease transmission in colder months.
Collapse
Affiliation(s)
- Gerardo Chowell
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, 31 Center Dr, MSC 2220, Bethesda 20892-2220, Maryland, USA
- Mathematical, Computational & Modeling Sciences Center, School of Human Evolution and Social Change, Arizona State University, 900 S. Cady Mall, Tempe 85287-2402, Arizona, USA
| | - Lone Simonsen
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, 31 Center Dr, MSC 2220, Bethesda 20892-2220, Maryland, USA
- Department of Global Health, School of Public Health and Health Services, George Washington University, 2175 K Street, Washington, DC 20037, USA
| | - Sherry Towers
- Mathematical, Computational & Modeling Sciences Center, School of Human Evolution and Social Change, Arizona State University, 900 S. Cady Mall, Tempe 85287-2402, Arizona, USA
| | - Mark A Miller
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, 31 Center Dr, MSC 2220, Bethesda 20892-2220, Maryland, USA
| | - Cécile Viboud
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, 31 Center Dr, MSC 2220, Bethesda 20892-2220, Maryland, USA
| |
Collapse
|
226
|
Abstract
The threat of a virulent, highly transmissible pandemic virus has motivated an escalating research effort to identify the transmissible genotypes of animal viruses that cross over into the human population (animal–human transmission) and sustain human–human transmission. In addition to the pursuit of the viral genotype, a greater understanding of the host-virus phenotype of infectiousness, transmissibility and susceptibility will be required. This review examines experimental animal transmission of influenza for insights into human influenza transmission. Transmission is viewed as sequential steps that the virus must pass critical thresholds to achieve transmission and ultimately survival in the human host. In particular, a quantitative understanding in animal models of viral replication efficiency, airway viral load, exhaled viral aerosol load, environmental virus survival and host susceptibility will likely yield important insights. Computational modeling will enhance animal model data, as well as guide the use of pandemic mitigation strategies.
Collapse
Affiliation(s)
- Frederick Koster
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA and The Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| |
Collapse
|
227
|
Wu A, Su C, Wang D, Peng Y, Liu M, Hua S, Li T, Gao G, Tang H, Chen J, Liu X, Shu Y, Peng D, Jiang T. Sequential Reassortments Underlie Diverse Influenza H7N9 Genotypes in China. Cell Host Microbe 2013; 14:446-52. [DOI: 10.1016/j.chom.2013.09.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 08/28/2013] [Accepted: 08/31/2013] [Indexed: 11/28/2022]
|
228
|
Determinants of virulence of influenza A virus. Eur J Clin Microbiol Infect Dis 2013; 33:479-90. [PMID: 24078062 DOI: 10.1007/s10096-013-1984-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/10/2013] [Indexed: 01/08/2023]
Abstract
Influenza A viruses cause yearly seasonal epidemics and occasional global pandemics in humans. In the last century, four human influenza A virus pandemics have occurred. Occasionally, influenza A viruses that circulate in other species cross the species barrier and infect humans. Virus reassortment (i.e. mixing of gene segments of multiple viruses) and the accumulation of mutations contribute to the emergence of new influenza A virus variants. Fortunately, most of these variants do not have the ability to spread among humans and subsequently cause a pandemic. In this review, we focus on the threat of animal influenza A viruses which have shown the ability to infect humans. In addition, genetic factors which could alter the virulence of influenza A viruses are discussed. The identification and characterisation of these factors may provide insights into genetic traits which change virulence and help us to understand which genetic determinants are of importance for the pandemic potential of animal influenza A viruses.
Collapse
|
229
|
Fonville JM, Burke DF, Lewis NS, Katzelnick LC, Russell CA. Quantifying the fitness advantage of polymerase substitutions in Influenza A/H7N9 viruses during adaptation to humans. PLoS One 2013; 8:e76047. [PMID: 24086684 PMCID: PMC3785442 DOI: 10.1371/journal.pone.0076047] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 08/22/2013] [Indexed: 01/15/2023] Open
Abstract
Adaptation of zoonotic influenza viruses towards efficient human-to-human transmissibility is a substantial public health concern. The recently emerged A/H7N9 influenza viruses in China provide an opportunity for quantitative studies of host-adaptation, as human-adaptive substitutions in the PB2 gene of the virus have been found in all sequenced human strains, while these substitutions have not been detected in any non-human A/H7N9 sequences. Given the currently available information, this observation suggests that the human-adaptive PB2 substitution might confer a fitness advantage to the virus in these human hosts that allows it to rise to proportions detectable by consensus sequencing over the course of a single human infection. We use a mathematical model of within-host virus evolution to estimate the fitness advantage required for a substitution to reach predominance in a single infection as a function of the duration of infection and the fraction of mutant present in the virus population that initially infects a human. The modeling results provide an estimate of the lower bound for the fitness advantage of this adaptive substitution in the currently sequenced A/H7N9 viruses. This framework can be more generally used to quantitatively estimate fitness advantages of adaptive substitutions based on the within-host prevalence of mutations. Such estimates are critical for models of cross-species transmission and host-adaptation of influenza virus infections.
Collapse
Affiliation(s)
- Judith M. Fonville
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - David F. Burke
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Nicola S. Lewis
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Leah C. Katzelnick
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Colin A. Russell
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
230
|
Belser JA, Gustin KM, Pearce MB, Maines TR, Zeng H, Pappas C, Sun X, Carney PJ, Villanueva JM, Stevens J, Katz JM, Tumpey TM. Pathogenesis and transmission of avian influenza A (H7N9) virus in ferrets and mice. Nature 2013; 501:556-9. [PMID: 23842497 PMCID: PMC7094885 DOI: 10.1038/nature12391] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 06/20/2013] [Indexed: 11/09/2022]
Abstract
On 29 March 2013, the Chinese Center for Disease Control and Prevention confirmed the first reported case of human infection with an avian influenza A(H7N9) virus. The recent human infections with H7N9 virus, totalling over 130 cases with 39 fatalities to date, have been characterized by severe pulmonary disease and acute respiratory distress syndrome (ARDS). This is concerning because H7 viruses have typically been associated with ocular disease in humans, rather than severe respiratory disease. This recent outbreak underscores the need to better understand the pathogenesis and transmission of these viruses in mammals. Here we assess the ability of A/Anhui/1/2013 and A/Shanghai/1/2013 (H7N9) viruses, isolated from fatal human cases, to cause disease in mice and ferrets and to transmit to naive animals. Both H7N9 viruses replicated to higher titre in human airway epithelial cells and in the respiratory tract of ferrets compared to a seasonal H3N2 virus. Moreover, the H7N9 viruses showed greater infectivity and lethality in mice compared to genetically related H7N9 and H9N2 viruses. The H7N9 viruses were readily transmitted to naive ferrets through direct contact but, unlike the seasonal H3N2 virus, did not transmit readily by respiratory droplets. The lack of efficient respiratory droplet transmission was corroborated by low receptor-binding specificity for human-like α2,6-linked sialosides. Our results indicate that H7N9 viruses have the capacity for efficient replication in mammals and human airway cells and highlight the need for continued public health surveillance of this emerging virus.
Collapse
Affiliation(s)
- Jessica A. Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Kortney M. Gustin
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Melissa B. Pearce
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Taronna R. Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Hui Zeng
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Claudia Pappas
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Xiangjie Sun
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Paul J. Carney
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Julie M. Villanueva
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - James Stevens
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Jacqueline M. Katz
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| | - Terrence M. Tumpey
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30333 Georgia USA
| |
Collapse
|
231
|
Abstract
Recently, novel H7N9 influenza viruses have caused an unprecedented outbreak in humans. Pigs are an important intermediate host for influenza; thus, we assessed the replication ability of three human H7N9 viruses (A/Anhui/1/2013, A/Shanghai/1/2013, A/Shanghai/2/2013) in swine tissue explants. All viruses tested replicated efficiently in explants from tracheas and bronchi, with limited replication in alveolar cells. Swine respiratory tissue explants can serve as an efficient model for screening replication potential of newly emerging H7N9 viruses.
Collapse
|
232
|
Belser JA, Tumpey TM. Tropism of H7N9 influenza viruses in the human respiratory tract. THE LANCET. RESPIRATORY MEDICINE 2013; 1:501-2. [PMID: 24461601 PMCID: PMC11299738 DOI: 10.1016/s2213-2600(13)70161-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | - Terrence M Tumpey
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| |
Collapse
|
233
|
Chan MCW, Chan RWY, Chan LLY, Mok CKP, Hui KPY, Fong JHM, Tao KP, Poon LLM, Nicholls JM, Guan Y, Peiris JSM. Tropism and innate host responses of a novel avian influenza A H7N9 virus: an analysis of ex-vivo and in-vitro cultures of the human respiratory tract. THE LANCET. RESPIRATORY MEDICINE 2013; 1:534-42. [PMID: 24461614 PMCID: PMC7164816 DOI: 10.1016/s2213-2600(13)70138-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Since March, 2013, an avian-origin influenza A H7N9 virus has caused severe pneumonia in China. The aim of this study was to investigate the pathogenesis of this new virus in human beings. METHODS We obtained ex-vivo cultures of the human bronchus, lung, nasopharynx, and tonsil and in-vitro cultures of primary human alveolar epithelial cells and peripheral blood monocyte-derived macrophages. We compared virus tropism and induction of proinflammatory cytokine responses of two human influenza A H7N9 virus isolates, A/Shanghai/1/2013 and A/Shanghai/2/2013; a highly pathogenic avian influenza H5N1 virus; the highly pathogenic avian influenza H7N7 virus that infected human beings in the Netherlands in 2003; the 2009 pandemic influenza H1N1 virus, and a low pathogenic duck H7N9 virus that was genetically different to the human disease causing A H7N9 viruses. FINDINGS Both human H7N9 viruses replicated efficiently in human bronchus and lung ex-vivo cultures, whereas duck/H7N9 virus failed to replicate in either. Both human A H7N9 viruses infected both ciliated and non-ciliated human bronchial epithelial cells and replicated to higher titres than did H5N1 (p<0.0001 to 0.0046) and A/Shanghai/1/2013 replicated to higher titres than did H7N7 (p=0.0002-0.01). Both human A H7N9 viruses predominantly infected type II alveolar epithelial cells and alveolar macrophages in the human lung and replicated to higher titres than did H5N1 (p<0.0001 to 0.0078); A/Shanghai/1/2013 replicated to higher titres than did H1N1 (p=0.0052-0.05) and H7N7 (p=0.0031-0.0151). Human H7N9 viruses were less potent inducers of proinflammatory cytokines compared with H5N1 virus. INTERPRETATION Collectively, the results suggest that the novel H7N9 viruses are better adapted to infect and replicate in the human conducting and lower airways than are other avian influenza viruses, including H5N1, and pose an important pandemic threat. FUNDING Area of Excellence Scheme of the University Grants Committee (AoE/M-12/96), Hong Kong Special Administrative Region.
Collapse
Affiliation(s)
- Michael C W Chan
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Renee W Y Chan
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Louisa L Y Chan
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chris K P Mok
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kenrie P Y Hui
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Joanne H M Fong
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kin P Tao
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Leo L M Poon
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - John M Nicholls
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Y Guan
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Joint Influenza Research Centre (SUMC/HKU), Shantou University Medical College/Hong Kong University, Shantou, China
| | - J S Malik Peiris
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
234
|
To KKW, Chan JFW, Chen H, Li L, Yuen KY. The emergence of influenza A H7N9 in human beings 16 years after influenza A H5N1: a tale of two cities. THE LANCET. INFECTIOUS DISEASES 2013; 13:809-21. [PMID: 23969217 PMCID: PMC7158959 DOI: 10.1016/s1473-3099(13)70167-1] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Infection with either influenza A H5N1 virus in 1997 or avian influenza A H7N9 virus in 2013 caused severe pneumonia that did not respond to typical or atypical antimicrobial treatment, and resulted in high mortality. Both viruses are reassortants with internal genes derived from avian influenza A H9N2 viruses that circulate in Asian poultry. Both viruses have genetic markers of mammalian adaptation in their haemagglutinin and polymerase PB2 subunits, which enhanced binding to human-type receptors and improved replication in mammals, respectively. Hong Kong (affected by H5N1 in 1997) and Shanghai (affected by H7N9 in 2013) are two rapidly flourishing cosmopolitan megacities that were increasing in human population and poultry consumption before the outbreaks. Both cities are located along the avian migratory route at the Pearl River delta and Yangtze River delta. Whether the widespread use of the H5N1 vaccine in east Asia-with suboptimum biosecurity measures in live poultry markets and farms-predisposed to the emergence of H7N9 or other virus subtypes needs further investigation. Why H7N9 seems to be more readily transmitted from poultry to people than H5N1 is still unclear.
Collapse
Affiliation(s)
- Kelvin KW To
- State Key Laboratory for Emerging Infectious Diseases, Research Centre of Infection and Immunology, Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jasper FW Chan
- State Key Laboratory for Emerging Infectious Diseases, Research Centre of Infection and Immunology, Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Honglin Chen
- State Key Laboratory for Emerging Infectious Diseases, Research Centre of Infection and Immunology, Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 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
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Research Centre of Infection and Immunology, Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
235
|
Prosser DJ, Hungerford LL, Erwin RM, Ottinger MA, Takekawa JY, Ellis EC. Mapping avian influenza transmission risk at the interface of domestic poultry and wild birds. Front Public Health 2013; 1:28. [PMID: 24350197 PMCID: PMC3854848 DOI: 10.3389/fpubh.2013.00028] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/06/2013] [Indexed: 12/02/2022] Open
Abstract
Emergence of avian influenza viruses with high lethality to humans, such as the currently circulating highly pathogenic A(H5N1) (emerged in 1996) and A(H7N9) cause serious concern for the global economic and public health sectors. Understanding the spatial and temporal interface between wild and domestic populations, from which these viruses emerge, is fundamental to taking action. This information, however, is rarely considered in influenza risk models, partly due to a lack of data. We aim to identify areas of high transmission risk between domestic poultry and wild waterfowl in China, the epicenter of both viruses. Two levels of models were developed: one that predicts hotspots of novel virus emergence between domestic and wild birds, and one that incorporates H5N1 risk factors, for which input data exists. Models were produced at 1 and 30 km spatial resolution, and two temporal seasons. Patterns of risk varied between seasons with higher risk in the northeast, central-east, and western regions of China during spring and summer, and in the central and southeastern regions during winter. Monte-Carlo uncertainty analyses indicated varying levels of model confidence, with lowest errors in the densely populated regions of eastern and southern China. Applications and limitations of the models are discussed within.
Collapse
Affiliation(s)
- Diann J Prosser
- Patuxent Wildlife Research Center, U.S. Geological Survey , Beltsville, MD , USA ; Marine Estuarine Environmental Sciences, University of Maryland , College Park, MD , USA
| | - Laura L Hungerford
- Marine Estuarine Environmental Sciences, University of Maryland , College Park, MD , USA ; University of Maryland School of Medicine , Baltimore, MD , USA
| | - R Michael Erwin
- Patuxent Wildlife Research Center, U.S. Geological Survey , Charlottesville, VA , USA
| | - Mary Ann Ottinger
- Department of Animal and Avian Sciences, University of Maryland , College Park, MD , USA
| | - John Y Takekawa
- Western Ecological Research Center, U.S. Geological Survey , Vallejo, CA , USA
| | - Erle C Ellis
- Department of Geography and Environmental Systems, University of Maryland Baltimore County , Baltimore, MD , USA
| |
Collapse
|
236
|
Xu L, Bao L, Deng W, Dong L, Zhu H, Chen T, Lv Q, Li F, Yuan J, Xiang Z, Gao K, Xu Y, Huang L, Li Y, Liu J, Yao Y, Yu P, Li X, Huang W, Zhao X, Lan Y, Guo J, Yong W, Wei Q, Chen H, Zhang L, Qin C. Novel avian-origin human influenza A(H7N9) can be transmitted between ferrets via respiratory droplets. J Infect Dis 2013; 209:551-6. [PMID: 23990570 DOI: 10.1093/infdis/jit474] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The outbreak of human infections caused by novel avian-origin influenza A(H7N9) in China since March 2013 underscores the need to better understand the pathogenicity and transmissibility of these viruses in mammals. In a ferret model, the pathogenicity of influenza A(H7N9) was found to be less than that of an influenza A(H5N1) strain but comparable to that of 2009 pandemic influenza A(H1N1), based on the clinical signs, mortality, virus dissemination, and results of histopathologic analyses. Influenza A(H7N9) could replicate in the upper and lower respiratory tract, the heart, the liver, and the olfactory bulb. It is worth noting that influenza A(H7N9) exhibited a low level of transmission between ferrets via respiratory droplets. There were 4 mutations in the virus isolated from the contact ferret: D678Y in the gene encoding PB2, R157K in the gene encoding hemagglutinin (H3 numbering), I109T in the gene encoding nucleoprotein, and T10I in the gene encoding neuraminidase. These data emphasized that avian-origin influenza A(H7N9) can be transmitted between mammals, highlighting its potential for human-to-human transmissibility.
Collapse
Affiliation(s)
- Lili Xu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Key Laboratory of Human Disease Comparative Medicine, Ministry of Health
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Chan RWY, Karamanska R, Van Poucke S, Van Reeth K, Chan IWW, Chan MCW, Dell A, Peiris JSM, Haslam SM, Guan Y, Nicholls JM. Infection of swine ex vivo tissues with avian viruses including H7N9 and correlation with glycomic analysis. Influenza Other Respir Viruses 2013; 7:1269-82. [PMID: 24001121 PMCID: PMC4114536 DOI: 10.1111/irv.12144] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2013] [Indexed: 01/21/2023] Open
Abstract
Objectives Swine have been regarded as intermediate hosts in the spread of influenza from birds to humans but studies of the sialylated glycans that comprise their respiratory tract have not been extensively studied in the past. This study analyzed the sialylated N‐glycan and O‐glycan profile of swine trachea and lung and correlated this with ex‐vivo infection of swine explants with avian influenza viruses. Sample Lungs and tracheal samples were obtained from normal farm and laboratory raised swine and used for ex vivo infection as well as mass spectrometric analysis. Infection of the ex vivo tissues used high pathogenic and low pathogenic avian viruses including the novel H7N9 virus that emerged in China in early 2013. Main outcome measures Assessment of successful replication was determined by TCID50 as well as virus immunohistochemistry. The N‐glycan and O‐glycan profiles were measured by MALDI‐TOF and sialylated linkages were determined by sialidase treatment. Lectin binding histochemistry was also performed on formalin fixed tissue samples with positive binding detected by chromogen staining. Results The swine respiratory tract glycans differed from the human respiratory tact glycans in two main areas. There was a greater abundance of Gal‐α‐Gal linkages resulting in a relative decrease in sialylated glycans. The swine respiratory tract also had a greater proportion of glycans containing Neu5Gc and Siaα2‐6 glycans than the human respiratory tract. Infection with avian viruses was confined primarily to lung bronchioles rather than trachea and parenchyma. Conclusions In contrast to previous studies we found that there was not as much expression of Siaα2‐3 glycans on the surface of the trachea. Infection of Siaα2‐3 binding avian viruses was restricted to the lower respiratory tract bronchioles. This finding may diminish the ability of the swine to act as an intermediary in the transmission of avian viruses to humans.
Collapse
Affiliation(s)
- Renee W Y Chan
- Department of Pathology, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre of Influenza Research, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Ramos I, Krammer F, Hai R, Aguilera D, Bernal-Rubio D, Steel J, García-Sastre A, Fernandez-Sesma A. H7N9 influenza viruses interact preferentially with α2,3-linked sialic acids and bind weakly to α2,6-linked sialic acids. J Gen Virol 2013; 94:2417-2423. [PMID: 23950563 DOI: 10.1099/vir.0.056184-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The recent human outbreak of H7N9 avian influenza A virus has caused worldwide concerns. Receptor binding specificity is critical for viral pathogenicity, and still not thoroughly studied for this emerging virus. Here, we evaluated the receptor specificity of the haemagglutinin (HA) of two human H7N9 isolates (A/Shanghai/1/13 and A/Anhui/1/13) through a solid-phase binding assay and a flow cytometry-based assay. In addition, we compared it with those from several HAs from human and avian influenza viruses. We observed that the HAs from the novel H7 isolates strongly interacted with α2,3-linked sialic acids. Importantly, they also showed low levels of binding to α2,6-linked sialic acids, but significantly higher than other avian H7s.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| | - Rong Hai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| | - Domingo Aguilera
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| | - Dabeiba Bernal-Rubio
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| | - John Steel
- Department of Microbiology and Immunology, Emory University School of Medicine, GA 30322, USA
| | - Adolfo García-Sastre
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| | - Ana Fernandez-Sesma
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue New York, NY 10029, USA
| |
Collapse
|
239
|
Abstract
Every year, influenza causes seasonal epidemics and is responsible for increased morbidity and mortality worldwide. In general, clinical signs include fever, respiratory symptoms and myalgia. Human influenza viruses include influenza viruses A/H1N1, A/H3N2, A/H1N1pdm09, influenza virus B and influenza virus C. Wild waterfowl are the natural reservoir of influenza A virus. On a sporadic basis novel viruses can emerge from the zoonotic reservoir and cross the species barrier. In such events clinical presentation in humans can range from mild (e.g. conjunctivitis) to severe pneumonia and acute respiratory distress syndrome (ARDS). In early 2013, a novel avian influenza A virus H7N9 emerged in China and the clinical presentation included bilateral pneumonia with ARDS and a high case fatality rate. In this article a summary is given on diagnosis, therapy and prophylaxis of influenza and on future developments.
Collapse
Affiliation(s)
- M. Panning
- Department für Medizinische Mikrobiologie und Hygiene, Institut für Virologie, Universitätsklinikum Freiburg, Hermann-Herder Str. 11, 70104 Freiburg, Deutschland
| |
Collapse
|
240
|
Langlois RA, Albrecht RA, Kimble B, Sutton T, Shapiro JS, Finch C, Angel M, Chua MA, Gonzalez-Reiche AS, Xu K, Perez D, García-Sastre A, tenOever BR. MicroRNA-based strategy to mitigate the risk of gain-of-function influenza studies. Nat Biotechnol 2013; 31:844-847. [PMID: 23934176 PMCID: PMC3808852 DOI: 10.1038/nbt.2666] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/19/2013] [Indexed: 01/01/2023]
Abstract
A strategy exploiting species-specific miRNA expression may provide another layer of biosafety in gain-of-function influenza experiments. Recent gain-of-function studies in influenza A virus H5N1 strains revealed that as few as three-amino-acid changes in the hemagglutinin protein confer the capacity for viral transmission between ferrets1,2. As transmission between ferrets is considered a surrogate indicator of transmissibility between humans, these studies raised concerns about the risks of gain-of-function influenza A virus research. Here we present an approach to strengthen the biosafety of gain-of-function influenza experiments. We exploit species-specific endogenous small RNAs to restrict influenza A virus tropism. In particular, we found that the microRNA miR-192 was expressed in primary human respiratory tract epithelial cells as well as in mouse lungs but absent from the ferret respiratory tract. Incorporation of miR-192 target sites into influenza A virus did not prevent influenza replication and transmissibility in ferrets, but did attenuate influenza pathogenicity in mice. This molecular biocontainment approach should be applicable beyond influenza A virus to minimize the risk of experiments involving other pathogenic viruses.
Collapse
Affiliation(s)
- Ryan A Langlois
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Global Health and Emerging Pathogens Institute, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Global Health and Emerging Pathogens Institute, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Brian Kimble
- Department of Veterinary Medicine, University of Maryland, College Park College Park, MD 20742
| | - Troy Sutton
- Department of Veterinary Medicine, University of Maryland, College Park College Park, MD 20742
| | - Jillian S Shapiro
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Courtney Finch
- Department of Veterinary Medicine, University of Maryland, College Park College Park, MD 20742
| | - Matthew Angel
- Department of Veterinary Medicine, University of Maryland, College Park College Park, MD 20742
| | - Mark A Chua
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | | | - Kemin Xu
- Department of Veterinary Medicine, University of Maryland, College Park College Park, MD 20742
| | - Daniel Perez
- Department of Veterinary Medicine, University of Maryland, College Park College Park, MD 20742
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Global Health and Emerging Pathogens Institute, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Benjamin R tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Global Health and Emerging Pathogens Institute, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
241
|
Xu L, Bao L, Deng W, Zhu H, Chen T, Lv Q, Li F, Yuan J, Xiang Z, Gao K, Xu Y, Huang L, Li Y, Liu J, Yao Y, Yu P, Yong W, Wei Q, Zhang L, Qin C. The mouse and ferret models for studying the novel avian-origin human influenza A (H7N9) virus. Virol J 2013; 10:253. [PMID: 23927489 PMCID: PMC3750532 DOI: 10.1186/1743-422x-10-253] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 08/07/2013] [Indexed: 02/06/2023] Open
Abstract
Background The current study was conducted to establish animal models (including mouse and ferret) for the novel avian-origin H7N9 influenza virus. Findings A/Anhui/1/2013 (H7N9) virus was administered by intranasal instillation to groups of mice and ferrets, and animals developed typical clinical signs including body weight loss (mice and ferrets), ruffled fur (mice), sneezing (ferrets), and death (mice). Peak virus shedding from respiratory tract was observed on 2 days post inoculation (d.p.i.) for mice and 3–5 d.p.i. for ferrets. Virus could also be detected in brain, liver, spleen, kidney, and intestine from inoculated mice, and in heart, liver, and olfactory bulb from inoculated ferrets. The inoculation of H7N9 could elicit seroconversion titers up to 1280 in ferrets and 160 in mice. Leukopenia, significantly reduced lymphocytes but increased neutrophils were also observed in mouse and ferret models. Conclusions The mouse and ferret model enables detailed studies of the pathogenesis of this illness and lay the foundation for drug or vaccine evaluation.
Collapse
Affiliation(s)
- Lili Xu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comptive Medicine Center, Peking Union Medical Collage (PUMC), Key Laboratory of Human Disease Comptive Medicine, Ministry of Health, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Richard M, Schrauwen EJA, de Graaf M, Bestebroer TM, Spronken MIJ, van Boheemen S, de Meulder D, Lexmond P, Linster M, Herfst S, Smith DJ, van den Brand JM, Burke DF, Kuiken T, Rimmelzwaan GF, Osterhaus ADME, Fouchier RAM. Limited airborne transmission of H7N9 influenza A virus between ferrets. Nature 2013; 501:560-3. [PMID: 23925116 PMCID: PMC3819191 DOI: 10.1038/nature12476] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/17/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Mathilde Richard
- Department of Viroscience, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Zhu Z, Yang Y, Feng Y, Shi B, Chen L, Zheng Y, Tian D, Song Z, Xu C, Qin B, Zhang X, Guan W, Liu F, Yang T, Yang H, Zeng D, Zhou W, Hu Y, Zhou X. Infection of inbred BALB/c and C57BL/6 and outbred Institute of Cancer Research mice with the emerging H7N9 avian influenza virus. Emerg Microbes Infect 2013; 2:e50. [PMID: 26038485 PMCID: PMC3821289 DOI: 10.1038/emi.2013.50] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/03/2013] [Accepted: 07/07/2013] [Indexed: 01/21/2023]
Abstract
A new avian-origin influenza virus A (H7N9) recently crossed the species barrier and infected humans; therefore, there is an urgent need to establish mammalian animal models for studying the pathogenic mechanism of this strain and the immunological response. In this study, we attempted to develop mouse models of H7N9 infection because mice are traditionally the most convenient models for studying influenza viruses. We showed that the novel A (H7N9) virus isolated from a patient could infect inbred BALB/c and C57BL/6 mice as well as outbred Institute of Cancer Research (ICR) mice. The amount of bodyweight lost showed differences at 7 days post infection (d.p.i.) (BALB/c mice 30%, C57BL/6 and ICR mice approximately 20%), and the lung indexes were increased both at 3 d.p.i. and at 7 d.p.i.. Immunohistochemistry demonstrated the existence of the H7N9 viruses in the lungs of the infected mice, and these findings were verified by quantitative real-time polymerase chain reaction (RT-PCR) and 50% tissue culture infectious dose (TCID50) detection at 3 d.p.i. and 7 d.p.i.. Histopathological changes occurred in the infected lungs, including pulmonary interstitial inflammatory lesions, pulmonary oedema and haemorrhages. Furthermore, because the most clinically severe cases were in elderly patients, we analysed the H7N9 infections in both young and old ICR mice. The old ICR mice showed more severe infections with more bodyweight lost and a higher lung index than the young ICR mice. Compared with the young ICR mice, the old mice showed a delayed clearance of the H7N9 virus and higher inflammation in the lungs. Thus, old ICR mice could partially mimic the more severe illness in elderly patients.
Collapse
Affiliation(s)
- Zhaoqin Zhu
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Yuqin Yang
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Yanling Feng
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Bisheng Shi
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Lixiang Chen
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Ye Zheng
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Di Tian
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Zhigang Song
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Chunhua Xu
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Boyin Qin
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Xiaonan Zhang
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Wencai Guan
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Fang Liu
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Tao Yang
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Hua Yang
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Dong Zeng
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Wenjiang Zhou
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China
| | - Yunwen Hu
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China ; Key laboratory of Medical Molecular Virology of the Ministries of Education, School of Basic Medical Science, Fudan Univeristy, Shanghai 200032, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Fudan University , Shanghai 201508, China ; Key laboratory of Medical Molecular Virology of the Ministries of Education, School of Basic Medical Science, Fudan Univeristy, Shanghai 200032, China
| |
Collapse
|
244
|
Yiu Lai K, Wing Yiu Ng G, Fai Wong K, Fan Ngai Hung I, Kam Fai Hong J, Fan Cheng F, Kwok Cheung Chan J. Human H7N9 avian influenza virus infection: a review and pandemic risk assessment. Emerg Microbes Infect 2013; 2:e48. [PMID: 26038484 PMCID: PMC3824111 DOI: 10.1038/emi.2013.48] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 06/12/2013] [Accepted: 06/18/2013] [Indexed: 12/20/2022]
Abstract
China is undergoing a recent outbreak of a novel H7N9 avian influenza virus (nH7N9) infection that has thus far involved 132 human patients, including 37 deaths. The nH7N9 virus is a reassortant virus originating from the H7N3, H7N9 and H9N2 avian influenza viruses. nH7N9 isolated from humans contains features related to adaptation to humans, including a Q226L mutation in the hemagglutinin cleavage site and E627K and D701N mutations in the PB2 protein. Live poultry markets provide an environment for the emergence, spread and maintenance of nH7N9 as well as for the selection of mutants that facilitate nH7N9 binding to and replication in the human upper respiratory tract. Innate immune suppression conferred by the internal genes of H9N2 may contribute to the virulence of nH7N9. The quail may serve as the intermediate host during the adaptation of avian influenza viruses from domestic waterfowl to gallinaceous poultry, such as chickens and related terrestrial-based species, due to the selection of viral mutants with a short neuraminidase stalk. Infections in chickens, common quails, red-legged partridges and turkeys may select for mutants with human receptor specificity. Infection in Ratitae species may lead to the selection of PB2-E627K and PB2-D701N mutants and the conversion of nH7N9 to a highly pathogenic avian influenza virus.
Collapse
Affiliation(s)
- Kang Yiu Lai
- Department of Intensive Care, Queen Elizabeth Hospital , Hong Kong, China
| | - George Wing Yiu Ng
- Department of Intensive Care, Queen Elizabeth Hospital , Hong Kong, China
| | - Kit Fai Wong
- Department of Pathology, Queen Elizabeth Hospital , Hong Kong, China
| | | | | | - Fanny Fan Cheng
- Department of Medicine, Queen Elizabeth Hospital , Hong Kong, China
| | | |
Collapse
|
245
|
Qi X, Qian YH, Bao CJ, Guo XL, Cui LB, Tang FY, Ji H, Huang Y, Cai PQ, Lu B, Xu K, Shi C, Zhu FC, Zhou MH, Wang H. Probable person to person transmission of novel avian influenza A (H7N9) virus in Eastern China, 2013: epidemiological investigation. BMJ 2013; 347:f4752. [PMID: 23920350 PMCID: PMC3805478 DOI: 10.1136/bmj.f4752] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To determine whether the novel avian influenza H7N9 virus can transmit from person to person and its efficiency. DESIGN Epidemiological investigations conducted after a family cluster of two patients with avian H7N9 in March 2013. SETTING Wuxi, Eastern China. PARTICIPANTS Two patients, their close contacts, and relevant environments. Samples from the patients and environments were collected and tested by real time reverse transcriptase-polymerase chain reaction (rRT-PCR), viral culture, and haemagglutination inhibition assay. Any contacts who became ill had samples tested for avian H7N9 by rRT-PCR. Paired serum samples were obtained from contacts for serological testing by haemagglutination inhibition assays. MAIN OUTCOMES MEASURES Clinical data, history of exposure before the onset of illnesses, and results of laboratory testing of pathogens and further analysis of sequences and phylogenetic tree to isolated strains. RESULTS The index patient became ill five to six days after his last exposure to poultry. The second patient, his daughter aged 32, who provided unprotected bedside care in the hospital, had no known exposure to poultry. She developed symptoms six days after her last contact with her father. Two strains were isolated successfully from the two patients. Genome sequence and analyses of phylogenetic trees showed that both viruses were almost genetically identical. Forty three close contacts of both patients were identified. One had mild illness but had negative results for avian H7N9 by rRT-PCR. All 43 close contacts tested negative for haemagglutination inhibition antibodies specific for avian H7N9. CONCLUSIONS The infection of the daughter probably resulted from contact with her father (the index patient) during unprotected exposure, suggesting that in this cluster the virus was able to transmit from person to person. The transmissibility was limited and non-sustainable.
Collapse
Affiliation(s)
- Xian Qi
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Province Center for Disease Control and Prevention, Nanjing, Jiangsu 210009, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Philippidis A. Gene therapy briefs. Hum Gene Ther 2013; 24:641-3. [PMID: 23876018 DOI: 10.1089/hum.2013.2508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
247
|
|
248
|
Characterization of H7N9 influenza A viruses isolated from humans. Nature 2013; 501:551-5. [PMID: 23842494 PMCID: PMC3891892 DOI: 10.1038/nature12392] [Citation(s) in RCA: 335] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 06/20/2013] [Indexed: 01/20/2023]
Abstract
Avian influenza A viruses rarely infect humans; however, when human infection and subsequent human-to-human transmission occurs, worldwide outbreaks (pandemics) can result. The recent sporadic infections of humans in China with a previously unrecognized avian influenza A virus of the H7N9 subtype (A(H7N9)) have caused concern owing to the appreciable case fatality rate associated with these infections (more than 25%), potential instances of human-to-human transmission, and the lack of pre-existing immunity among humans to viruses of this subtype. Here we characterize two early human A(H7N9) isolates, A/Anhui/1/2013 (H7N9) and A/Shanghai/1/2013 (H7N9); hereafter referred to as Anhui/1 and Shanghai/1, respectively. In mice, Anhui/1 and Shanghai/1 were more pathogenic than a control avian H7N9 virus (A/duck/Gunma/466/2011 (H7N9); Dk/GM466) and a representative pandemic 2009 H1N1 virus (A/California/4/2009 (H1N1pdm09); CA04). Anhui/1, Shanghai/1 and Dk/GM466 replicated well in the nasal turbinates of ferrets. In nonhuman primates, Anhui/1 and Dk/GM466 replicated efficiently in the upper and lower respiratory tracts, whereas the replicative ability of conventional human influenza viruses is typically restricted to the upper respiratory tract of infected primates. By contrast, Anhui/1 did not replicate well in miniature pigs after intranasal inoculation. Critically, Anhui/1 transmitted through respiratory droplets in one of three pairs of ferrets. Glycan arrays showed that Anhui/1, Shanghai/1 and A/Hangzhou/1/2013 (H7N9) (a third human A(H7N9) virus tested in this assay) bind to human virus-type receptors, a property that may be critical for virus transmissibility in ferrets. Anhui/1 was found to be less sensitive in mice to neuraminidase inhibitors than a pandemic H1N1 2009 virus, although both viruses were equally susceptible to an experimental antiviral polymerase inhibitor. The robust replicative ability in mice, ferrets and nonhuman primates and the limited transmissibility in ferrets of Anhui/1 suggest that A(H7N9) viruses have pandemic potential.
Collapse
|
249
|
Abstract
ABSTRACT The ongoing H7N9 influenza epizootic in China once again presents us questions about the origin of pandemics and how to recognize them in early stages of development. Over the past ~135 years, H7 influenza viruses have neither caused pandemics nor been recognized as having undergone human adaptation. Yet several unusual properties of these viruses, including their poultry epizootic potential, mammalian adaptation, and atypical clinical syndromes in rarely infected humans, suggest that they may be different from other avian influenza viruses, thus questioning any assurance that the likelihood of human adaptation is low. At the same time, the H7N9 epizootic provides an opportunity to learn more about the mammalian/human adaptational capabilities of avian influenza viruses and challenges us to integrate virologic and public health research and surveillance at the animal-human interface.
Collapse
|