Cargando…

Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30

BACKGROUND: The genetic basis for avian to mammalian host switching in influenza A virus is largely unknown. The human A/HK/156/1997 (H5N1) virus that transmitted from poultry possesses NS1 gene mutations F103L + M106I that are virulence determinants in the mouse model of pneumonia; however their in...

Descripción completa

Detalles Bibliográficos
Autores principales: Dankar, Samar K, Miranda, Elena, Forbes, Nicole E, Pelchat, Martin, Tavassoli, Ali, Selman, Mohammed, Ping, Jihui, Jia, Jianjun, Brown, Earl G
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3733596/
https://www.ncbi.nlm.nih.gov/pubmed/23886034
http://dx.doi.org/10.1186/1743-422X-10-243
_version_ 1782279367141883904
author Dankar, Samar K
Miranda, Elena
Forbes, Nicole E
Pelchat, Martin
Tavassoli, Ali
Selman, Mohammed
Ping, Jihui
Jia, Jianjun
Brown, Earl G
author_facet Dankar, Samar K
Miranda, Elena
Forbes, Nicole E
Pelchat, Martin
Tavassoli, Ali
Selman, Mohammed
Ping, Jihui
Jia, Jianjun
Brown, Earl G
author_sort Dankar, Samar K
collection PubMed
description BACKGROUND: The genetic basis for avian to mammalian host switching in influenza A virus is largely unknown. The human A/HK/156/1997 (H5N1) virus that transmitted from poultry possesses NS1 gene mutations F103L + M106I that are virulence determinants in the mouse model of pneumonia; however their individual roles have not been determined. The emergent A/Shanghai/patient1/2013(H7N9)-like viruses also possess these mutations which may contribute to their virulence and ability to switch species. METHODS: NS1 mutant viruses were constructed by reverse genetics and site directed mutagenesis on human and mouse-adapted backbones. Mouse infections assessed virulence, virus yield, tissue infection, and IFN induction. NS1 protein properties were assessed for subcellular distribution, IFN antagonism (mouse and human), CPSF30 and RIG-I domain binding, host transcription (microarray); and the natural prevalence of 103L and 106I mutants was assessed. RESULTS: Each of the F103L and M106I mutations contributes additively to virulence to reduce the lethal dose by >800 and >3,200 fold respectively by mediating alveolar tissue infection with >100 fold increased infectious yields. The 106I NS1 mutant lost CPSF binding but the 103L mutant maintained binding that correlated with an increased general decrease in host gene expression in human but not mouse cells. Each mutation positively modulated the inhibition of IFN induction in mouse cells and activation of the IFN-β promoter in human cells but not in combination in human cells indicating negative epistasis. Each of the F103L and M106I mutations restored a defect in cytoplasmic localization of H5N1 NS1 in mouse cells. Human H1N1 and H3N2 NS1 proteins bound to the CARD, helicase and RD RIG-I domains, whereas the H5N1 NS1 with the same consensus 103F and 106M mutations did not bind these domains, which was totally or partially restored by the M106I or F103L mutations respectively. CONCLUSIONS: The F103L and M106I mutations in the H5N1 NS1 protein each increased IFN antagonism and mediated interstitial pneumonia in mice that was associated with increased cytoplasmic localization and altered host factor binding. These mutations may contribute to the ability of previous HPAI H5N1 and recent LPAI H7N9 and H6N1 (NS1-103L+106M) viruses to switch hosts and cause disease in humans.
format Online
Article
Text
id pubmed-3733596
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-37335962013-08-06 Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30 Dankar, Samar K Miranda, Elena Forbes, Nicole E Pelchat, Martin Tavassoli, Ali Selman, Mohammed Ping, Jihui Jia, Jianjun Brown, Earl G Virol J Research BACKGROUND: The genetic basis for avian to mammalian host switching in influenza A virus is largely unknown. The human A/HK/156/1997 (H5N1) virus that transmitted from poultry possesses NS1 gene mutations F103L + M106I that are virulence determinants in the mouse model of pneumonia; however their individual roles have not been determined. The emergent A/Shanghai/patient1/2013(H7N9)-like viruses also possess these mutations which may contribute to their virulence and ability to switch species. METHODS: NS1 mutant viruses were constructed by reverse genetics and site directed mutagenesis on human and mouse-adapted backbones. Mouse infections assessed virulence, virus yield, tissue infection, and IFN induction. NS1 protein properties were assessed for subcellular distribution, IFN antagonism (mouse and human), CPSF30 and RIG-I domain binding, host transcription (microarray); and the natural prevalence of 103L and 106I mutants was assessed. RESULTS: Each of the F103L and M106I mutations contributes additively to virulence to reduce the lethal dose by >800 and >3,200 fold respectively by mediating alveolar tissue infection with >100 fold increased infectious yields. The 106I NS1 mutant lost CPSF binding but the 103L mutant maintained binding that correlated with an increased general decrease in host gene expression in human but not mouse cells. Each mutation positively modulated the inhibition of IFN induction in mouse cells and activation of the IFN-β promoter in human cells but not in combination in human cells indicating negative epistasis. Each of the F103L and M106I mutations restored a defect in cytoplasmic localization of H5N1 NS1 in mouse cells. Human H1N1 and H3N2 NS1 proteins bound to the CARD, helicase and RD RIG-I domains, whereas the H5N1 NS1 with the same consensus 103F and 106M mutations did not bind these domains, which was totally or partially restored by the M106I or F103L mutations respectively. CONCLUSIONS: The F103L and M106I mutations in the H5N1 NS1 protein each increased IFN antagonism and mediated interstitial pneumonia in mice that was associated with increased cytoplasmic localization and altered host factor binding. These mutations may contribute to the ability of previous HPAI H5N1 and recent LPAI H7N9 and H6N1 (NS1-103L+106M) viruses to switch hosts and cause disease in humans. BioMed Central 2013-07-25 /pmc/articles/PMC3733596/ /pubmed/23886034 http://dx.doi.org/10.1186/1743-422X-10-243 Text en Copyright ©2013 Dankar et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research
Dankar, Samar K
Miranda, Elena
Forbes, Nicole E
Pelchat, Martin
Tavassoli, Ali
Selman, Mohammed
Ping, Jihui
Jia, Jianjun
Brown, Earl G
Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30
title Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30
title_full Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30
title_fullStr Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30
title_full_unstemmed Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30
title_short Influenza A/Hong Kong/156/1997(H5N1) virus NS1 gene mutations F103L and M106I both increase IFN antagonism, virulence and cytoplasmic localization but differ in binding to RIG-I and CPSF30
title_sort influenza a/hong kong/156/1997(h5n1) virus ns1 gene mutations f103l and m106i both increase ifn antagonism, virulence and cytoplasmic localization but differ in binding to rig-i and cpsf30
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3733596/
https://www.ncbi.nlm.nih.gov/pubmed/23886034
http://dx.doi.org/10.1186/1743-422X-10-243
work_keys_str_mv AT dankarsamark influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT mirandaelena influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT forbesnicolee influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT pelchatmartin influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT tavassoliali influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT selmanmohammed influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT pingjihui influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT jiajianjun influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30
AT brownearlg influenzaahongkong1561997h5n1virusns1genemutationsf103landm106ibothincreaseifnantagonismvirulenceandcytoplasmiclocalizationbutdifferinbindingtorigiandcpsf30