Advertisement

H7N9 Adaptation

Puzzling and alarming reports of an outbreak in early 2013 of human infections by a low-pathogenicity avian influenza virus has rocked the poultry industry in central eastern China and brought fears of initiating a human pandemic. Over 130 human cases have been reported with 37 deaths until closure of poultry markets accompanied a near-cessation of human case reports. From surveillance sampling of >10,000 isolates obtained during April 2013, Zhang et al. (p. 410, published online 18 July) took 37 isolates of avian origin H7N9 and compared them to human H7N9 isolates. The majority of H7N9 isolates came from live poultry markets, although some originated in pigeons. Sequence analysis indicated that the chicken isolates had retained the avian characteristics at sites on the influenza genes for PB2 and the surface hemagglutinin HA, where adaptive mutations have been observed before. Sequence analysis also showed a higher variability in the internal genes than in HA and neuraminidase NA. By using glycan arrays, it was shown that avian and human isolates bound to human, but also to some extent to avian, receptors. As expected, the virus replicated well in chickens without causing disease, whereas in mice only the human isolates were highly pathogenic. The human virus, but not the avian, transmitted between ferrets through the air.

Abstract

A newly emerged H7N9 virus has caused 132 human infections with 37 deaths in China since 18 February 2013. Control measures in H7N9 virus–positive live poultry markets have reduced the number of infections; however, the character of the virus, including its pandemic potential, remains largely unknown. We systematically analyzed H7N9 viruses isolated from birds and humans. The viruses were genetically closely related and bound to human airway receptors; some also maintained the ability to bind to avian airway receptors. The viruses isolated from birds were nonpathogenic in chickens, ducks, and mice; however, the viruses isolated from humans caused up to 30% body weight loss in mice. Most importantly, one virus isolated from humans was highly transmissible in ferrets by respiratory droplet. Our findings indicate nothing to reduce the concern that these viruses can transmit between humans.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Supplementary Text
Figs. S1 to S4
Tables S1 to S4
References

Resources

File (zhang.sm.pdf)

Reference and Notes

1
Tong S., Li Y., Rivailler P., Conrardy C., Castillo D. A., Chen L. M., Recuenco S., Ellison J. A., Davis C. T., York I. A., Turmelle A. S., Moran D., Rogers S., Shi M., Tao Y., Weil M. R., Tang K., Rowe L. A., Sammons S., Xu X., Frace M., Lindblade K. A., Cox N. J., Anderson L. J., Rupprecht C. E., Donis R. O., A distinct lineage of influenza A virus from bats. Proc. Natl. Acad. Sci. U.S.A. 109, 4269–4274 (2012).
2
World Health Organisation, Number of confirmed human cases of avian influenza A(H7N9) reported to WHO, www.who.int/influenza/human_animal_interface/influenza_h7n9/08_ReportWebH7N9Number.pdf (May 2013).
3
See supplementary materials on Science Online.
4
Shi J., Deng G. H., Liu P. H., Zhou J. P., Guan L. Z., Li W. H., Li X. Y., Guo J., Wang G. J., Fan J., Wang J. L., Li Y. Y., Jiang Y. P., Liu L. L., Tian G. B., Li C. J., Chen H. L., Isolation and characterization of H7N9 viruses from live poultry markets—Implication of the source of current H7N9 infection in humans. Chin. Sci. Bull. 58, 1857–1863 (2013).
5
Gao R., Cao B., Hu Y., Feng Z., Wang D., Hu W., Chen J., Jie Z., Qiu H., Xu K., Xu X., Lu H., Zhu W., Gao Z., Xiang N., Shen Y., He Z., Gu Y., Zhang Z., Yang Y., Zhao X., Zhou L., Li X., Zou S., Zhang Y., Li X., Yang L., Guo J., Dong J., Li Q., Dong L., Zhu Y., Bai T., Wang S., Hao P., Yang W., Zhang Y., Han J., Yu H., Li D., Gao G. F., Wu G., Wang Y., Yuan Z., Shu Y., Human infection with a novel avian-origin influenza A (H7N9) virus. N. Engl. J. Med. 368, 1888–1897 (2013).
6
Chen Y., Liang W., Yang S., Wu N., Gao H., Sheng J., Yao H., Wo J., Fang Q., Cui D., Li Y., Yao X., Zhang Y., Wu H., Zheng S., Diao H., Xia S., Zhang Y., Chan K. H., Tsoi H. W., Teng J. L., Song W., Wang P., Lau S. Y., Zheng M., Chan J. F., To K. K., Chen H., Li L., Yuen K. Y., Human infections with the emerging avian influenza A H7N9 virus from wet market poultry: Clinical analysis and characterisation of viral genome. Lancet 381, 1916–1925 (2013).
7
Single-letter abbreviations for the amino acid residues are as follows: A, Ala; C, Cys; D, Asp; E, Glu; F, Phe; G, Gly; H, His; I, Ile; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q, Gln; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr.
8
Subbarao E. K., London W., Murphy B. R., A single amino acid in the PB2 gene of influenza A virus is a determinant of host range. J. Virol. 67, 1761–1764 (1993).
9
Li Z., Chen H., Jiao P., Deng G., Tian G., Li Y., Hoffmann E., Webster R. G., Matsuoka Y., Yu K., Molecular basis of replication of duck H5N1 influenza viruses in a mammalian mouse model. J. Virol. 79, 12058–12064 (2005).
10
Gao Y., Zhang Y., Shinya K., Deng G., Jiang Y., Li Z., Guan Y., Tian G., Li Y., Shi J., Liu L., Zeng X., Bu Z., Xia X., Kawaoka Y., Chen H., Identification of amino acids in HA and PB2 critical for the transmission of H5N1 avian influenza viruses in a mammalian host. PLoS Pathog. 5, e1000709 (2009).
11
Hatta M., Gao P., Halfmann P., Kawaoka Y., Molecular basis for high virulence of Hong Kong H5N1 influenza A viruses. Science 293, 1840–1842 (2001).
12
Glaser L., Stevens J., Zamarin D., Wilson I. A., García-Sastre A., Tumpey T. M., Basler C. F., Taubenberger J. K., Palese P., A single amino acid substitution in 1918 influenza virus hemagglutinin changes receptor binding specificity. J. Virol. 79, 11533–11536 (2005).
13
Matrosovich M., Tuzikov A., Bovin N., Gambaryan A., Klimov A., Castrucci M. R., Donatelli I., Kawaoka Y., Early alterations of the receptor-binding properties of H1, H2, and H3 avian influenza virus hemagglutinins after their introduction into mammals. J. Virol. 74, 8502–8512 (2000).
14
Imai M., Watanabe T., Hatta M., Das S. C., Ozawa M., Shinya K., Zhong G., Hanson A., Katsura H., Watanabe S., Li C., Kawakami E., Yamada S., Kiso M., Suzuki Y., Maher E. A., Neumann G., Kawaoka Y., Experimental adaptation of an influenza H5 HA confers respiratory droplet transmission to a reassortant H5 HA/H1N1 virus in ferrets. Nature 486, 420–428 (2012).
15
Herfst S., Schrauwen E. J., Linster M., Chutinimitkul S., de Wit E., Munster V. J., Sorrell E. M., Bestebroer T. M., Burke D. F., Smith D. J., Rimmelzwaan G. F., Osterhaus A. D., Fouchier R. A., Airborne transmission of influenza A/H5N1 virus between ferrets. Science 336, 1534–1541 (2012).
16
Vines A., Wells K., Matrosovich M., Castrucci M. R., Ito T., Kawaoka Y., The role of influenza A virus hemagglutinin residues 226 and 228 in receptor specificity and host range restriction. J. Virol. 72, 7626–7631 (1998).
17
Rogers G. N., Paulson J. C., Receptor determinants of human and animal influenza virus isolates: differences in receptor specificity of the H3 hemagglutinin based on species of origin. Virology 127, 361–373 (1983).
18
Zhang Y., Zhang Q., Kong H., Jiang Y., Gao Y., Deng G., Shi J., Tian G., Liu L., Liu J., Guan Y., Bu Z., Chen H., H5N1 hybrid viruses bearing 2009/H1N1 virus genes transmit in guinea pigs by respiratory droplet. Science 340, 1459–1463 (2013).
19
Kageyama T., Fujisaki S., Takashita E., Xu H., Yamada S., Uchida Y., Neumann G., Saito T., Kawaoka Y., Tashiro M., Genetic analysis of novel avian A(H7N9) influenza viruses isolated from patients in China, February to April 2013. Euro Surveill. 18, 20453 (2013).
20
Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (Office International des Epizooties, Paris, 2011).
21
Li Q., Zhou L., Zhou M., Chen Z., Li F., Wu H., Xiang N., Chen E., Tang F., Wang D., Meng L., Hong Z., Tu W., Cao Y., Li L., Ding F., Liu B., Wang M., Xie R., Gao R., Li X., Bai T., Zou S., He J., Hu J., Xu Y., Chai C., Wang S., Gao Y., Jin L., Zhang Y., Luo H., Yu H., Gao L., Pang X., Liu G., Shu Y., Yang W., Uyeki T. M., Wang Y., Wu F., Feng Z., Preliminary report: Epidemiology of the avian influenza A (H7N9) outbreak in China. N. Engl. J. Med. 130424140638006 (2013).
22
Lu X., Tumpey T. M., Morken T., Zaki S. R., Cox N. J., Katz J. M., A mouse model for the evaluation of pathogenesis and immunity to influenza A (H5N1) viruses isolated from humans. J. Virol. 73, 5903–5911 (1999).
23
Li Y., Shi J., Zhong G., Deng G., Tian G., Ge J., Zeng X., Song J., Zhao D., Liu L., Jiang Y., Guan Y., Bu Z., Chen H., Continued evolution of H5N1 influenza viruses in wild birds, domestic poultry, and humans in China from 2004 to 2009. J. Virol. 84, 8389–8397 (2010).
24
Zhang Y., Zhang Q., Gao Y., He X., Kong H., Jiang Y., Guan Y., Xia X., Shu Y., Kawaoka Y., Bu Z., Chen H., Key molecular factors in hemagglutinin and PB2 contribute to efficient transmission of the 2009 H1N1 pandemic influenza virus. J. Virol. 86, 9666–9674 (2012).
25
Lowen A. C., Mubareka S., Tumpey T. M., García-Sastre A., Palese P., The guinea pig as a transmission model for human influenza viruses. Proc. Natl. Acad. Sci. U.S.A. 103, 9988–9992 (2006).
26
Seibert C. W., Kaminski M., Philipp J., Rubbenstroth D., Albrecht R. A., Schwalm F., Stertz S., Medina R. A., Kochs G., García-Sastre A., Staeheli P., Palese P., Oseltamivir-resistant variants of the 2009 pandemic H1N1 influenza A virus are not attenuated in the guinea pig and ferret transmission models. J. Virol. 84, 11219–11226 (2010).
27
Tumpey T. M., Maines T. R., Van Hoeven N., Glaser L., Solórzano A., Pappas C., Cox N. J., Swayne D. E., Palese P., Katz J. M., García-Sastre A., A two-amino acid change in the hemagglutinin of the 1918 influenza virus abolishes transmission. Science 315, 655–659 (2007).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 341 | Issue 6144
26 July 2013

Article versions

You are viewing the most recent version of this article.

Submission history

Received: 15 May 2013
Accepted: 2 July 2013
Published in print: 26 July 2013

Permissions

Request permissions for this article.

Acknowledgments

We thank S. Watson for editing the manuscript, Y. Shu from the China Centers for Disease Control and Prevention for providing the H7N9 viruses isolated from humans, and the Consortium for Functional Glycomics (Scripps Research Institute, Department of Molecular Biology, La Jolla, CA) for providing the glycans. This work was supported by the Ministry of Agriculture (CARS-42-G08) and by the Ministry of Science and Technology (KJYJ-2013-01-01 and 2012ZX10004214). Virus sequence data from this study were deposited in GenBank with the accession numbers CY146905 to CY147200 and in Global Initiative on Sharing Avian Influenza Data with the accession numbers EPI440678 to 440701 and EIP457614 to 457885.

Authors

Affiliations

Qianyi Zhang*
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730030, People’s Republic of China.
Jianzhong Shi*
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Guohua Deng*
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Jing Guo*
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Xianying Zeng*
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Xijun He
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Huihui Kong
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Chunyang Gu
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730030, People’s Republic of China.
Xuyong Li
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Jinxiong Liu
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Guojun Wang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730030, People’s Republic of China.
Yan Chen
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Liling Liu
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Libin Liang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Yuanyuan Li
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Jun Fan
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Jinliang Wang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Wenhui Li
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Lizheng Guan
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Qimeng Li
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730030, People’s Republic of China.
Huanliang Yang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Pucheng Chen
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Li Jiang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Yuntao Guan
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Xiaoguang Xin
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Yongping Jiang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Guobin Tian
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Xiurong Wang
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Chuanling Qiao
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Chengjun Li
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Zhigao Bu
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
Hualan Chen [email protected]
State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, People’s Republic of China.
College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730030, People’s Republic of China.

Notes

*
These authors contributed equally to this work.
Corresponding author. E-mail: [email protected]

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Genetic characterization and pathogenicity of H7N9 highly pathogenic avian influenza viruses isolated from South China in 2017, Frontiers in Microbiology, 14, (2023).https://doi.org/10.3389/fmicb.2023.1105529
    Crossref
  2. Characterization of an H7N9 Influenza Virus Isolated from Camels in Inner Mongolia, China, Microbiology Spectrum, (2023).https://doi.org/10.1128/spectrum.01798-22
    Crossref
  3. Avian H7N9 influenza viruses are evolutionarily constrained by stochastic processes during replication and transmission in mammals, Virus Evolution, 9, 1, (2023).https://doi.org/10.1093/ve/vead004
    Crossref
  4. Enhanced pathogenicity and transmissibility of H9N2 avian influenza virus in mammals by hemagglutinin mutations combined with PB2-627K, Virologica Sinica, 38, 1, (47-55), (2023).https://doi.org/10.1016/j.virs.2022.09.006
    Crossref
  5. H10Nx avian influenza viruses detected in wild birds in China pose potential threat to mammals, One Health, 16, (100515), (2023).https://doi.org/10.1016/j.onehlt.2023.100515
    Crossref
  6. Advances in deciphering the interactions between viral proteins of influenza A virus and host cellular proteins, Cell Insight, 2, 2, (100079), (2023).https://doi.org/10.1016/j.cellin.2023.100079
    Crossref
  7. Genetic analysis and biological characterization of H10N3 influenza A viruses isolated in China from 2014 to 2021, Journal of Medical Virology, 95, 2, (2023).https://doi.org/10.1002/jmv.28476
    Crossref
  8. H1N1 Influenza A Virus Protein NS2 Inhibits Innate Immune Response by Targeting IRF7, Viruses, 14, 11, (2411), (2022).https://doi.org/10.3390/v14112411
    Crossref
  9. Evolution of Swine Influenza Virus H3N2 in Vaccinated and Nonvaccinated Pigs after Previous Natural H1N1 Infection, Viruses, 14, 9, (2008), (2022).https://doi.org/10.3390/v14092008
    Crossref
  10. ARNT Inhibits H5N1 Influenza A Virus Replication by Interacting with the PA Protein, Viruses, 14, 7, (1347), (2022).https://doi.org/10.3390/v14071347
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media