Advertisement
No access
Research Article
Host-Pathogen Interactions

Inhibition of Autophagy Ameliorates Acute Lung Injury Caused by Avian Influenza A H5N1 Infection

Science Signaling
21 Feb 2012
Vol 5, Issue 212
p. ra16

Preventing H5N1 from Damaging Lungs

The higher mortality rate of the H5N1 strain of avian influenza compared to that of the seasonal H1N1 virus is attributed to the more severe lung damage caused by the H5N1 strain. Sun et al. found that lung tissue from an individual infected with H5N1 contained many autophagosomes and that mice infected with H5N1 had greater numbers of autophagosomes in lung tissue than did mice infected with the H1N1 strain. In addition, the H5N1 virus stimulated autophagic signaling in mouse epithelial cells to a greater extent than did the H1N1 virus. Pharmacological inhibition of autophagic signaling or knockdown of components of the autophagy pathway in H5N1-infected mice resulted in less severe lung damage, increased survival rate, and decreased mortality. These findings suggest that targeting the autophagy pathway might provide therapeutic targets in treating H5N1 infection in humans.

Abstract

The threat of a new influenza pandemic has existed since 1997, when the highly pathogenic H5N1 strain of avian influenza A virus infected humans in Hong Kong and spread across Asia, where it continued to infect poultry and people. The human mortality rate of H5N1 infection is about 60%, whereas that of seasonal H1N1 infection is less than 0.1%. The high mortality rate associated with H5N1 infection is predominantly a result of respiratory failure caused by acute lung injury; however, how viral infection contributes to this disease pathology is unclear. Here, we used electron microscopy to show the accumulation of autophagosomes in H5N1-infected lungs from a human cadaver and mice, as well as in infected A549 human epithelial lung cells. We also showed that H5N1, but not seasonal H1N1, induced autophagic cell death in alveolar epithelial cells through a pathway involving the kinase Akt, the tumor suppressor protein TSC2, and the mammalian target of rapamycin. Additionally, we suggest that the hemagglutinin protein of H5N1 may be responsible for stimulating autophagy. When applied prophylactically, reagents that blocked virus-induced autophagic signaling substantially increased the survival rate of mice and substantially ameliorated the acute lung injury and mortality caused by H5N1 infection. We conclude that the autophagic cell death of alveolar epithelial cells likely plays a crucial role in the high mortality rate of H5N1 infection, and we suggest that autophagy-blocking agents might be useful as prophylactics and therapeutics against infection of humans by the H5N1 virus.

Get full access to this article

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

Supplementary Material

Summary

Fig. S1. Autophagy is induced by H5N1 infection of mouse lungs and A549 cells.
Fig. S2. LC3 puncta occur in H5N1-infected A549 cells and decrease in abundance upon treatment with TSC2-specific siRNA.
Fig. S3. Autophagic death of A549 cells induced by H5N1 infection is substantially rescued by Atg6-specific siRNA.
Fig. S4. The production of proinflammatory cytokines by H5N1 infection is not altered by Atg5-specific siRNA.
Fig. S5. The increased immune response induced by H5N1 infection is not altered by Atg5-specific siRNA.
Fig. S6. Mouse model of acute lung injury induced by infection with live H5N1 virus.
Fig. S7. Acute lung injury in mice induced by live H5N1 virus was ameliorated by Atg6-specific siRNA.
Fig. S8. Acute lung injury in mice induced by live H5N1 virus was not ameliorated by the selective inhibitors of apoptosis or necrosis.
Fig. S9. Mice infected by H1N1 virus and treated with Atg5-specific siRNA do not develop acute lung injury.
Fig. S10. Rag1 knockout mice develop acute lung injury induced by H5N1 virus.
Fig. S11. The HA protein of H5N1 virus may induce autophagy in human A549 cells.

Resources

File (5_ra16_sm.pdf)

References and Notes

1
Gambotto A., Barratt-Boyes S. M., de Jong M. D., Neumann G., Kawaoka Y., Human infection with highly pathogenic H5N1 influenza virus. Lancet 371, 1464–1475 (2008).
2
Bauer T. T., Ewig S., Rodloff A. C., Muller E. E., Acute respiratory distress syndrome and pneumonia: A comprehensive review of clinical data. Clin. Infect. Dis. 43, 748–756 (2006).
3
Yu H., Gao Z., Feng Z., Shu Y., Xiang N., Zhou L., Huai Y., Feng L., Peng Z., Li Z., Xu C., Li J., Hu C., Li Q., Xu X., Liu X., Liu Z., Xu L., Chen Y., Luo H., Wei L., Zhang X., Xin J., Guo J., Wang Q., Yuan Z., Zhang K., Zhang W., Yang J., Zhong X., Xia S., Li L., Cheng J., Ma E., He P., Lee S. S., Wang Y., Uyeki T. M., Yang W., Clinical characteristics of 26 human cases of highly pathogenic avian influenza A (H5N1) virus infection in China. PLoS One 3, e2985 (2008).
4
Thanh T. T., van Doorn H. R., de Jong M. D., Human H5N1 influenza: Current insight into pathogenesis. Int. J. Biochem. Cell Biol. 40, 2671–2674 (2008).
5
Wang H., Jiang C., Avian influenza H5N1: An update on molecular pathogenesis. Sci. China C Life Sci. 52, 459–463 (2009).
6
Wang H., Jiang C., Influenza A virus H5N1 entry into host cells is through clathrin-dependent endocytosis. Sci. China C Life Sci. 52, 464–469 (2009).
7
Neumann G., Chen H., Gao G. F., Shu Y., Kawaoka Y., H5N1 influenza viruses: Outbreaks and biological properties. Cell Res. 20, 51–61 (2010).
8
Imai Y., Kuba K., Neely G. G., Yaghubian-Malhami R., Perkmann T., van Loo G., Ermolaeva M., Veldhuizen R., Leung Y. H., Wang H., Liu H., Sun Y., Pasparakis M., Kopf M., Mech C., Bavari S., Peiris J. S., Slutsky A. S., Akira S., Hultqvist M., Holmdahl R., Nicholls J., Jiang C., Binder C. J., Penninger J. M., Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell 133, 235–249 (2008).
9
Korteweg C., Gu J., Pathology, molecular biology, and pathogenesis of avian influenza A (H5N1) infection in humans. Am. J. Pathol. 172, 1155–1170 (2008).
10
Mizushima N., Methods for monitoring autophagy. Int. J. Biochem. Cell Biol. 36, 2491–2502 (2004).
11
Kabeya Y., Mizushima N., Ueno T., Yamamoto A., Kirisako T., Noda T., Kominami E., Ohsumi Y., Yoshimori T., LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19, 5720–5728 (2000).
12
Yoshimura K., Shibata M., Koike M., Gotoh K., Fukaya M., Watanabe M., Uchiyama Y., Effects of RNA interference of Atg4B on the limited proteolysis of LC3 in PC12 cells and expression of Atg4B in various rat tissues. Autophagy 2, 200–208 (2006).
13
Mizushima N., Yoshimori T., How to interpret LC3 immunoblotting. Autophagy 3, 542–545 (2007).
14
Zhou Z., Jiang X., Liu D., Fan Z., Hu X., Yan J., Wang M., Gao G. F., Autophagy is involved in influenza A virus replication. Autophagy 5, 321–328 (2009).
15
Gannagé M., Dormann D., Albrecht R., Dengjel J., Torossi T., Rämer P. C., Lee M., Strowig T., Arrey F., Conenello G., Pypaert M., Andersen J., García-Sastre A., Münz C., Matrix protein 2 of influenza A virus blocks autophagosome fusion with lysosomes. Cell Host Microbe 6, 367–380 (2009).
16
Mizushima N., Autophagy: Process and function. Genes Dev. 21, 2861–2873 (2007).
17
Levine B., Cell biology: Autophagy and cancer. Nature 446, 745–747 (2007).
18
Baehrecke E. H., Autophagy: Dual roles in life and death? Nat. Rev. Mol. Cell Biol. 6, 505–510 (2005).
19
Seglen P. O., Gordon P. B., 3-Methyladenine: Specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc. Natl. Acad. Sci. U.S.A. 79, 1889–1892 (1982).
20
Blommaart E. F., Krause U., Schellens J. P., Vreeling-Sindelarova H., Meijer A. J., The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 inhibit autophagy in isolated rat hepatocytes. Eur. J. Biochem. 243, 240–246 (1997).
21
Pyo J. O., Jang M. H., Kwon Y. K., Lee H. J., Jun J. I., Woo H. N., Cho D. H., Choi B., Lee H., Kim J. H., Mizushima N., Oshumi Y., Jung Y. K., Essential roles of Atg5 and FADD in autophagic cell death: Dissection of autophagic cell death into vacuole formation and cell death. J. Biol. Chem. 280, 20722–20729 (2005).
22
Jounai N., Takeshita F., Kobiyama K., Sawano A., Miyawaki A., Xin K. Q., Ishii K. J., Kawai T., Akira S., Suzuki K., Okuda K., The Atg5–Atg12 conjugate associates with innate antiviral immune responses. Proc. Natl. Acad. Sci. U.S.A. 104, 14050–14055 (2007).
23
Díaz-Troya S., Pérez-Pérez M. E., Florencio F. J., Crespo J. L., The role of TOR in autophagy regulation from yeast to plants and mammals. Autophagy 4, 851–865 (2008).
24
Pattingre S., Espert L., Biard-Piechaczyk M., Codogno P., Regulation of macroautophagy by mTOR and Beclin 1 complexes. Biochimie 90, 313–323 (2008).
25
Deretic V., Autophagy in innate and adaptive immunity. Trends Immunol. 26, 523–528 (2005).
26
Klionsky D. J., Autophagy: From phenomenology to molecular understanding in less than a decade. Nat. Rev. Mol. Cell Biol. 8, 931–937 (2007).
27
Ehrhardt C., Wolff T., Pleschka S., Planz O., Beermann W., Bode J. G., Schmolke M., Ludwig S., Influenza A virus NS1 protein activates the PI3K/Akt pathway to mediate antiapoptotic signaling responses. J. Virol. 81, 3058–3067 (2007).
28
Cascone T., Morelli M. P., Morgillo F., Kim W. Y., Rodolico G., Pepe S., Tortora G., Berrino L., Lee H. Y., Heymach J. V., Ciardiello F., Synergistic anti-proliferative and pro-apoptotic activity of combined therapy with bortezomib, a proteasome inhibitor, with anti-epidermal growth factor receptor (EGFR) drugs in human cancer cells. J. Cell. Physiol. 216, 698–707 (2008).
29
Kuo C. T., Hsu M. J., Chen B. C., Chen C. C., Teng C. M., Pan S. L., Lin C. H., Denbinobin induces apoptosis in human lung adenocarcinoma cells via Akt inactivation, Bad activation, and mitochondrial dysfunction. Toxicol. Lett. 177, 48–58 (2008).
30
Shih Y. W., Chen P. S., Wu C. H., Jeng Y. F., Wang C. J., α-Chaconine-reduced metastasis involves a PI3K/Akt signaling pathway with downregulation of NF-κB in human lung adenocarcinoma A549 cells. J. Agric. Food Chem. 55, 11035–11043 (2007).
31
Preet A., Ganju R. K., Groopman J. E., Δ9-Tetrahydrocannabinol inhibits epithelial growth factor-induced lung cancer cell migration in vitro as well as its growth and metastasis in vivo. Oncogene 27, 339–346 (2008).
32
Morita M., Suyama H., Igishi T., Shigeoka Y., Kodani M., Hashimoto K., Takeda K., Sumikawa T., Shimizu E., Dexamethasone inhibits paclitaxel-induced cytotoxic activity through retinoblastoma protein dephosphorylation in non-small cell lung cancer cells. Int. J. Oncol. 30, 187–192 (2007).
33
Jang B. C., Lim K. J., Suh M. H., Park J. G., Suh S. I., Dexamethasone suppresses interleukin-1β-induced human β-defensin 2 mRNA expression: Involvement of p38 MAPK, JNK, MKP-1, and NF-κB transcriptional factor in A549 cells. FEMS Immunol. Med. Microbiol. 51, 171–184 (2007).
34
Zhang S., Xiao L., Zhou H., Yu Z., Chen H., Guo A., Jin M., Generation and characterization of an H5N1 avian influenza virus hemagglutinin glycoprotein pseudotyped lentivirus. J. Virol. Methods 154, 99–103 (2008).
35
Xu T., Qiao J., Zhao L., Wang G., He G., Li K., Tian Y., Gao M., Wang J., Wang H., Dong C., Acute respiratory distress syndrome induced by avian influenza A (H5N1) virus in mice. Am. J. Respir. Crit. Care Med. 174, 1011–1017 (2006).
36
Uiprasertkul M., Kitphati R., Puthavathana P., Kriwong R., Kongchanagul A., Ungchusak K., Angkasekwinai S., Chokephaibulkit K., Srisook K., Vanprapar N., Auewarakul P., Apoptosis and pathogenesis of avian influenza A (H5N1) virus in humans. Emerg. Infect. Dis. 13, 708–712 (2007).
37
Hu X. F., Liu Q. Z., Li C., Dong J., Zhou J. F., Wang M., Shu Y. L., Liu H. T., Liang M. F., Li D. X., The differential expression of the human lung carcinoma cells infected with high pathogenic avian influenza virus A/Anhui/1/2005 (H5N1). Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi 22, 180–182 (2008).
38
Li C., Liu H., Sun Y., Wang H., Guo F., Rao S., Deng J., Zhang Y., Miao Y., Guo C., Meng J., Chen X., Li L., Li D., Xu H., Wang H., Li B., Jiang C., PAMAM nanoparticles promote acute lung injury by inducing autophagic cell death through the Akt-TSC2-mTOR signaling pathway. J. Mol. Cell Biol. 1, 37–45 (2009).
39
Liu H. L., Zhang Y. L., Yang N., Zhang Y. X., Liu X. Q., Li C. G., Zhao Y., Wang Y. G., Zhang G. G., Yang P., Guo F., Sun Y., Jiang C. Y., A functionalized single-walled carbon nanotube-induced autophagic cell death in human lung cells through Akt-TSC2-mTOR signaling. Cell Death Dis. 2, e159 (2011).

(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 Signaling
Volume 5 | Issue 212
February 2012

Submission history

Received: 17 February 2011
Accepted: 2 February 2012

Permissions

Request permissions for this article.

Acknowledgments

We thank Z. Han, W. Xi, H. Wang, R. Lu, X. Chen, Z. Pan, R. Sheng, and X. Liu for technical support; H. Zhang and J. Ma for experiments performed in mouse embryonic fibroblast cells with formulated chicken inactivated virus vaccines; and J. Penninger, L. Li, and H. Pickersgill for helpful discussions and for editing the manuscript. Funding: This work was supported by the National Natural Science Foundation of China (30625013, 30623009, and 81000764), the Ministry of Science and Technology of China (2009CB522105), and the Ministry of Health (2009ZX10004-308). D.L. acknowledges support by the Science and Technology Commission of Shanghai Municipality (07pj14096). Author contributions: Y. Sun, C.L., X.J., Z.Z., H.W., S.R., F.G., W.N., Y.Z., Y.Y., J.T., and C.Z. performed the experiments; Y. Shu and R.G. provided H5N1-infected human lung tissue; H. Liu, P.Y., K.L., and S.W. constructed the H5-expressing cell line and expressed and purified the H5 protein; H. Lu, X.L., and L.T. prepared live virus; J.S. took EM photos of human tissue; X.G. provided Rag1 knockout mice; X.T., Y.Q., and K.-F.X. provided normal human lung tissue; D.L. provided helpful ideas; C.J. and N.J. designed the experiments and analyzed the data; and C.J. wrote the manuscript. Competing interests: C.J., S.R., H. Liu, F.G., H.W., Y. Sun, and C.L. have filed for a patent in China for the use of autophagy inhibitors in the prevention or treatment of acute lung injury. The file number: PCT/CN2010/072331.

Authors

Affiliations

Yang Sun*
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Chenggang Li*
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Yuelong Shu*
State Key Laboratory for Molecular Virology and Genetic Engineering, National Institute for Viral Disease Control and Prevention, China CDC, Changbai 155, Changping District, Beijing 102206, China.
Xiangwu Ju
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Zhen Zou
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Hongliang Wang
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Shuan Rao
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Feng Guo
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Haolin Liu
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Wenlong Nan
Genetic Engineering Laboratory, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130062, China.
Yan Zhao
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Yiwu Yan
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Jun Tang
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Chen Zhao
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Peng Yang
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Kangtai Liu
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Shunxin Wang
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.
Huijun Lu
Genetic Engineering Laboratory, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130062, China.
Xiao Li
Genetic Engineering Laboratory, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130062, China.
Lei Tan
Genetic Engineering Laboratory, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130062, China.
Rongbao Gao
State Key Laboratory for Molecular Virology and Genetic Engineering, National Institute for Viral Disease Control and Prevention, China CDC, Changbai 155, Changping District, Beijing 102206, China.
Jingdong Song
State Key Laboratory for Molecular Virology and Genetic Engineering, National Institute for Viral Disease Control and Prevention, China CDC, Changbai 155, Changping District, Beijing 102206, China.
Xiang Gao
MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, 22 Hankou Road, Nanjing 210093, China.
Xinlun Tian
Peking Union Medical College Hospital, No.1 Shuaifuyuan, Beijing 100005, China.
Yingzhi Qin
Peking Union Medical College Hospital, No.1 Shuaifuyuan, Beijing 100005, China.
Kai-Feng Xu
Peking Union Medical College Hospital, No.1 Shuaifuyuan, Beijing 100005, China.
Dangsheng Li
Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
Genetic Engineering Laboratory, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130062, China.
Chengyu Jiang [email protected]
State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing 100005, P. R. China.

Notes

*
These authors contributed equally to this work.
To whom correspondence should be addressed. E-mail: [email protected] (C.J.); [email protected] (N.J.)

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. Targeting influenza A virus by splicing inhibitor herboxidiene reveals the importance of subtype-specific signatures around splice sites, Journal of Biomedical Science, 30, 1, (2023).https://doi.org/10.1186/s12929-023-00897-4
    Crossref
  2. The role of transforming growth factor beta-1 protein in Escherichia coli secondary infection induced by H9N2 avian influenza virus in chickens, Microbial Pathogenesis, 175, (105983), (2023).https://doi.org/10.1016/j.micpath.2023.105983
    Crossref
  3. The Contribution of Viral Proteins to the Synergy of Influenza and Bacterial Co-Infection, Viruses, 14, 5, (1064), (2022).https://doi.org/10.3390/v14051064
    Crossref
  4. Effects of Glutamine Starvation on SHVV Replication by Quantitative Proteomics Analysis, Fishes, 7, 6, (315), (2022).https://doi.org/10.3390/fishes7060315
    Crossref
  5. Therapeutic Targeting of NF-κB in Acute Lung Injury: A Double-Edged Sword, Cells, 11, 20, (3317), (2022).https://doi.org/10.3390/cells11203317
    Crossref
  6. The Multi-Faceted Role of Autophagy During Animal Virus Infection, Frontiers in Cellular and Infection Microbiology, 12, (2022).https://doi.org/10.3389/fcimb.2022.858953
    Crossref
  7. circEXOC5 promotes acute lung injury through the PTBP1/Skp2/Runx2 axis to activate autophagy, Life Science Alliance, 6, 1, (e202201468), (2022).https://doi.org/10.26508/lsa.202201468
    Crossref
  8. Nonclassical autophagy activation pathways are essential for production of infectious Influenza A virus in vitro, Molecular Microbiology, 117, 2, (508-524), (2022).https://doi.org/10.1111/mmi.14865
    Crossref
  9. Avian influenza viruses suppress innate immunity by inducing trans-transcriptional readthrough via SSU72, Cellular & Molecular Immunology, 19, 6, (702-714), (2022).https://doi.org/10.1038/s41423-022-00843-8
    Crossref
  10. PKM2 regulates cigarette smoke-induced airway inflammation and epithelial-to-mesenchymal transition via modulating PINK1/Parkin-mediated mitophagy, Toxicology, 477, (153251), (2022).https://doi.org/10.1016/j.tox.2022.153251
    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.

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