Skip to main content
Top
Published in: Virology Journal 1/2018

Open Access 01-12-2018 | Research

Bat lung epithelial cells show greater host species-specific innate resistance than MDCK cells to human and avian influenza viruses

Authors: Tessa Slater, Isabella Eckerle, Kin-Chow Chang

Published in: Virology Journal | Issue 1/2018

Login to get access

Abstract

Background

With the recent discovery of novel H17N10 and H18N11 influenza viral RNA in bats and report on high frequency of avian H9 seroconversion in a species of free ranging bats, an important issue to address is the extent bats are susceptible to conventional avian and human influenza A viruses.

Method

To this end, three bat species (Eidolon helvum, Carollia perspicillata and Tadarida brasiliensis) of lung epithelial cells were separately infected with two avian and two human influenza viruses to determine their relative host innate immune resistance to infection.

Results

All three species of bat cells were more resistant than positive control Madin-Darby canine kidney (MDCK) cells to all four influenza viruses. TB1-Lu cells lacked sialic acid α2,6-Gal receptors and were most resistant among the three bat species. Interestingly, avian viruses were relatively more replication permissive in all three bat species of cells than with the use of human viruses which suggest that bats could potentially play a role in the ecology of avian influenza viruses. Chemical inhibition of the JAK-STAT pathway in bat cells had no effect on virus production suggesting that type I interferon signalling is not a major factor in resisting influenza virus infection.

Conclusion

Although all three species of bat cells are relatively more resistant to influenza virus infection than control MDCK cells, they are more permissive to avian than human viruses which suggest that bats could have a contributory role in the ecology of avian influenza viruses.
Literature
3.
go back to reference Bean AGD, Baker ML, Stewart CR, et al. Studying immunity to zoonotic diseases in the natural host - keeping it real. Nat Rev Immunol. 2013;13:851–61.CrossRefPubMed Bean AGD, Baker ML, Stewart CR, et al. Studying immunity to zoonotic diseases in the natural host - keeping it real. Nat Rev Immunol. 2013;13:851–61.CrossRefPubMed
4.
go back to reference Hoffman M, Müller MA, Drexler JF, et al. Differential sensitivity of bat cells to infection by enveloped RNA viruses: coronaviruses, paramyxoviruses, filoviruses and influenza viruses. PLoS One. 2013;8:e72942.CrossRef Hoffman M, Müller MA, Drexler JF, et al. Differential sensitivity of bat cells to infection by enveloped RNA viruses: coronaviruses, paramyxoviruses, filoviruses and influenza viruses. PLoS One. 2013;8:e72942.CrossRef
5.
go back to reference Sandekian V, Lim D, Prud'homme P, Lemay G. Transient high level mammalian reovirus replication in a bat epithelial cell line occurs without cytopathic effect. Virus Res. 2013;173:327–35.CrossRefPubMed Sandekian V, Lim D, Prud'homme P, Lemay G. Transient high level mammalian reovirus replication in a bat epithelial cell line occurs without cytopathic effect. Virus Res. 2013;173:327–35.CrossRefPubMed
6.
go back to reference Wu Y, Tefsen B, Shi Y, Gao GF. Bat-derived influenza-like viruses H17N10 and H18N11. Trends Microbiol. 2014;22:183–91.CrossRefPubMed Wu Y, Tefsen B, Shi Y, Gao GF. Bat-derived influenza-like viruses H17N10 and H18N11. Trends Microbiol. 2014;22:183–91.CrossRefPubMed
7.
go back to reference Juozapaitis M, Moreira EA, Mena I, et al. An infectious bat-derived chimeric influenza virus harbouring the entry machinery of an influenza a virus. Nat Commun. 2014;5:4448.CrossRefPubMedPubMedCentral Juozapaitis M, Moreira EA, Mena I, et al. An infectious bat-derived chimeric influenza virus harbouring the entry machinery of an influenza a virus. Nat Commun. 2014;5:4448.CrossRefPubMedPubMedCentral
8.
go back to reference Poole DS, Yu S, Cai Y, et al. Influenza a virus polymerase is a site for adaptive changes during experimental evolution in bat cells. J Virol. 2014;88:12572–85.CrossRefPubMedPubMedCentral Poole DS, Yu S, Cai Y, et al. Influenza a virus polymerase is a site for adaptive changes during experimental evolution in bat cells. J Virol. 2014;88:12572–85.CrossRefPubMedPubMedCentral
10.
go back to reference Dlugolenski D, Jones L, Tompkins SM, et al. Bat cells from Pteropus alecto are susceptible to influenza a virus infection and reassortment. Influenza Other Resp. 2013;7:900–3.CrossRef Dlugolenski D, Jones L, Tompkins SM, et al. Bat cells from Pteropus alecto are susceptible to influenza a virus infection and reassortment. Influenza Other Resp. 2013;7:900–3.CrossRef
11.
12.
13.
go back to reference Kuchipudi SV, Dunham SP, Nelli R, et al. Rapid death of duck cells infected with influenza: a potential mechanism for host resistance to H5N1. Immunol Cell Biol. 2012;90:116–23.CrossRefPubMed Kuchipudi SV, Dunham SP, Nelli R, et al. Rapid death of duck cells infected with influenza: a potential mechanism for host resistance to H5N1. Immunol Cell Biol. 2012;90:116–23.CrossRefPubMed
14.
go back to reference Nelli RK, Dunham SP, Kuchipudi SV, et al. Mammalian innate resistance to highly pathogenic avian influenza H5N1 virus infection is mediated through reduced proinflammation and infectious virus release. J Virol. 2012;86:9201–10.CrossRefPubMedPubMedCentral Nelli RK, Dunham SP, Kuchipudi SV, et al. Mammalian innate resistance to highly pathogenic avian influenza H5N1 virus infection is mediated through reduced proinflammation and infectious virus release. J Virol. 2012;86:9201–10.CrossRefPubMedPubMedCentral
15.
go back to reference Nelli RK, Kuchipudi SV, White GA, et al. Comparative distribution of human and avian type sialic acid influenza receptors in the pig. BMC Vet Res. 2010;6:4.CrossRefPubMedPubMedCentral Nelli RK, Kuchipudi SV, White GA, et al. Comparative distribution of human and avian type sialic acid influenza receptors in the pig. BMC Vet Res. 2010;6:4.CrossRefPubMedPubMedCentral
16.
go back to reference Ramos I, Bernal-Rubio D, Durham N, et al. Effects of receptor binding specificity of avian influenza virus on the human innate immune response. J Virol. 2011;85:4421–31.CrossRefPubMedPubMedCentral Ramos I, Bernal-Rubio D, Durham N, et al. Effects of receptor binding specificity of avian influenza virus on the human innate immune response. J Virol. 2011;85:4421–31.CrossRefPubMedPubMedCentral
17.
go back to reference Delgado-Ortega M, Melo S, Punyadarsaniya D, et al. Innate immune response to a H3N2 subtype swine influenza virus in newborn porcine trachea cells, alveolar macrophages, and precision-cut lung slices. Vet Res. 2014;45:42.CrossRefPubMedPubMedCentral Delgado-Ortega M, Melo S, Punyadarsaniya D, et al. Innate immune response to a H3N2 subtype swine influenza virus in newborn porcine trachea cells, alveolar macrophages, and precision-cut lung slices. Vet Res. 2014;45:42.CrossRefPubMedPubMedCentral
18.
go back to reference Gaush CR, Replication STF. Plaque assay of influenza virus in an established line of canine kidney cells. Appl Microbiol. 1968;16:588–94.PubMedPubMedCentral Gaush CR, Replication STF. Plaque assay of influenza virus in an established line of canine kidney cells. Appl Microbiol. 1968;16:588–94.PubMedPubMedCentral
19.
go back to reference Govorkova EA, Kodihalli S, Alymova IV, Fanget B, Webster RG. Growth and immunogenicity of influenza viruses cultivated in Vero or MDCK cells and in embryonated chicken eggs. Dev Biol Stand. 1999;98:39–51.PubMed Govorkova EA, Kodihalli S, Alymova IV, Fanget B, Webster RG. Growth and immunogenicity of influenza viruses cultivated in Vero or MDCK cells and in embryonated chicken eggs. Dev Biol Stand. 1999;98:39–51.PubMed
20.
go back to reference Panzer S. Transfection: viral and synthetic techniques converge; high-tech polymers leveraging viral biophysics can enhance the transfection of nucleic acids. Genet Engin Biotechnol News. 2014;34:20–1.CrossRef Panzer S. Transfection: viral and synthetic techniques converge; high-tech polymers leveraging viral biophysics can enhance the transfection of nucleic acids. Genet Engin Biotechnol News. 2014;34:20–1.CrossRef
21.
go back to reference Weis W, Brown JH, Cusack S, et al. Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Nature. 1988;333:426–31.CrossRefPubMed Weis W, Brown JH, Cusack S, et al. Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Nature. 1988;333:426–31.CrossRefPubMed
22.
go back to reference Gamblin SJ, Haire LF, Russell RJ, et al. The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science. 2004;303:1838–42.CrossRefPubMed Gamblin SJ, Haire LF, Russell RJ, et al. The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science. 2004;303:1838–42.CrossRefPubMed
23.
go back to reference Stevens J, Blixt O, Paulson JC, Wilson IA. Glycan microarray technologies: tools to survey host specificity of influenza viruses. Nat Rev Microbiol. 2006;4:857–64.CrossRefPubMed Stevens J, Blixt O, Paulson JC, Wilson IA. Glycan microarray technologies: tools to survey host specificity of influenza viruses. Nat Rev Microbiol. 2006;4:857–64.CrossRefPubMed
24.
go back to reference Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol. 2003;3(11):900.CrossRefPubMed Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol. 2003;3(11):900.CrossRefPubMed
25.
go back to reference Xie J, Li Y, Shen X, Goh G, Zhu Y, et al. Dampened STING-dependent interferon activation in bats. Host Cell Microbe. 2018;23:297–301.CrossRef Xie J, Li Y, Shen X, Goh G, Zhu Y, et al. Dampened STING-dependent interferon activation in bats. Host Cell Microbe. 2018;23:297–301.CrossRef
26.
go back to reference Pinto R, Herold S, Cakarova L, et al. Inhibition of influenza virus-induced NF-kappaB and Raf/MEK/ERK activation can reduce both virus titers and cytokine expression simultaneously in vitro and in vivo. Antivir Res. 2011;92:45–56.CrossRefPubMed Pinto R, Herold S, Cakarova L, et al. Inhibition of influenza virus-induced NF-kappaB and Raf/MEK/ERK activation can reduce both virus titers and cytokine expression simultaneously in vitro and in vivo. Antivir Res. 2011;92:45–56.CrossRefPubMed
27.
go back to reference Planz O. Development of cellular signaling pathway inhibitors as new antivirals against influenza. Antivir Res. 2013;98:457–68.CrossRefPubMed Planz O. Development of cellular signaling pathway inhibitors as new antivirals against influenza. Antivir Res. 2013;98:457–68.CrossRefPubMed
28.
29.
go back to reference Matikainen S, Sirén J, Tissari J, et al. Tumor necrosis factor alpha enhances influenza a virus-induced expression of antiviral cytokines by activating RIG-I gene expression. J Virol. 2006;80:3515–22.CrossRefPubMedPubMedCentral Matikainen S, Sirén J, Tissari J, et al. Tumor necrosis factor alpha enhances influenza a virus-induced expression of antiviral cytokines by activating RIG-I gene expression. J Virol. 2006;80:3515–22.CrossRefPubMedPubMedCentral
30.
go back to reference Bernasconi D, Amici C, La Frazia S, Ianaro A, Santoro MG. The IκB kinase is a key factor in triggering influenza a virus-induced inflammatory cytokine production in airway epithelial cells. J Biol Chem. 2005;280:24127–34.CrossRefPubMed Bernasconi D, Amici C, La Frazia S, Ianaro A, Santoro MG. The IκB kinase is a key factor in triggering influenza a virus-induced inflammatory cytokine production in airway epithelial cells. J Biol Chem. 2005;280:24127–34.CrossRefPubMed
Metadata
Title
Bat lung epithelial cells show greater host species-specific innate resistance than MDCK cells to human and avian influenza viruses
Authors
Tessa Slater
Isabella Eckerle
Kin-Chow Chang
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Virology Journal / Issue 1/2018
Electronic ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-018-0979-6

Other articles of this Issue 1/2018

Virology Journal 1/2018 Go to the issue