Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/112010
Title: An interferon regulated microRNA provides cell-intrinsec anti-viral immunity through multihit host-directed targeting of the sterol pathway
Author: Robertson, Kevin A.
Hsieh, Wei Yuan
Forster, Thorsten
Blanc, Mathieu
Lu, Hongjin
Crick, Peter J.
Yutuc, Eylan
Watterson, Steven
Martin, Kimberly
Griffiths, Samantha J.
Enright, Anton J.
Yamamoto, Mami
Pradeepa, Madapura M.
Lennox, Kimberly A.
Behlke, Mark A.
Talbot, Simon
Haas, Jürgen
Dolken, Lars
Griffiths, William J.
Wang, Yuqin
Angulo Aguado, Ana
Ghazal, Peter
Keywords: Interferó
Colesterol
Biosíntesi
Micro RNAs
Interferon
Cholesterol
Biosynthesis
MicroRNAs
Issue Date: 2016
Publisher: Public Library of Science (PLoS)
Abstract: In invertebrates, small interfering RNAs are at the vanguard of cell-autonomous antiviral immunity. In contrast, antiviral mechanisms initiated by interferon (IFN) signaling predominate in mammals. Whilst mammalian IFN-induced miRNA are known to inhibit specific viruses, it is not known whether host-directed microRNAs, downstream of IFN-signaling, have a role in mediating broad antiviral resistance. By performing an integrative, systematic, global analysis of RNA turnover utilizing 4-thiouridine labeling of newly transcribed RNA and pri/pre-miRNA in IFN-activated macrophages, we identify a new post-transcriptional viral defense mechanism mediated by miR-342-5p. On the basis of ChIP and site-directed promoter mutagenesis experiments, we find the synthesis of miR-342-5p is coupled to the antiviral IFN response via the IFN-induced transcription factor, IRF1. Strikingly, we find miR-342-5p targets mevalonate-sterol biosynthesis using a multihit mechanism suppressing the pathway at different functional levels: transcriptionally via SREBF2, post-transcriptionally via miR-33, and enzymatically via IDI1 and SC4MOL. Mass spectrometry-based lipidomics and enzymatic assays demonstrate the targeting mechanisms reduce intermediate sterol pathway metabolites and total cholesterol in macrophages. These results reveal a previously unrecognized mechanism by which IFN regulates the sterol pathway. The sterol pathway is known to be an integral part of the macrophage IFN antiviral response, and we show that miR-342-5p exerts broad antiviral effects against multiple, unrelated pathogenic viruses such Cytomegalovirus and Influenza A (H1N1). Metabolic rescue experiments confirm the specificity of these effects and demonstrate that unrelated viruses have differential mevalonate and sterol pathway requirements for their replication. This study, therefore, advances the general concept of broad antiviral defense through multihit targeting of a single host pathway.
Note: Reproducció del document publicat a: https://doi.org/10.1371/journal.pbio.1002364
It is part of: PLoS Biology, 2016, vol. 14, p. e1002364
URI: http://hdl.handle.net/2445/112010
Related resource: https://doi.org/10.1371/journal.pbio.1002364
ISSN: 1544-9173
Appears in Collections:Articles publicats en revistes (IDIBAPS: Institut d'investigacions Biomèdiques August Pi i Sunyer)
Articles publicats en revistes (Biomedicina)

Files in This Item:
File Description SizeFormat 
666611.pdf7.27 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons