5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      miR‐193b represses influenza A virus infection by inhibiting Wnt/β‐catenin signalling

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Due to an increasing emergence of new and drug‐resistant strains of the influenza A virus (IAV), developing novel measures to combat influenza is necessary. We have previously shown that inhibiting Wnt/β‐catenin pathway reduces IAV infection. In this study, we aimed to identify antiviral human microRNAs (miRNAs) that target the Wnt/β‐catenin signalling pathway. Using a miRNA expression library, we identified 85 miRNAs that up‐regulated and 20 miRNAs that down‐regulated the Wnt/β‐catenin signalling pathway. Fifteen miRNAs were validated to up‐regulate and five miRNAs to down‐regulate the pathway. Overexpression of four selected miRNAs (miR‐193b, miR‐548f‐1, miR‐1‐1, and miR‐509‐1) that down‐regulated the Wnt/β‐catenin signalling pathway reduced viral mRNA, protein levels in A/PR/8/34‐infected HEK293 cells, and progeny virus production. Overexpression of miR‐193b in lung epithelial A549 cells also resulted in decreases of A/PR/8/34 infection. Furthermore, miR‐193b inhibited the replication of various strains, including H1N1 (A/PR/8/34, A/WSN/33, A/Oklahoma/3052/09) and H3N2 (A/Oklahoma/309/2006), as determined by a viral reporter luciferase assay. Further studies revealed that β‐catenin was a target of miR‐193b, and β‐catenin rescued miR‐193b‐mediated suppression of IAV infection. miR‐193b induced G0/G1 cell cycle arrest and delayed vRNP nuclear import. Finally, adenovirus‐mediated gene transfer of miR‐193b to the lung reduced viral load in mice challenged by a sublethal dose of A/PR/8/34. Collectively, our findings suggest that miR‐193b represses IAV infection by inhibiting Wnt/β‐catenin signalling.

          Related collections

          Most cited references69

          • Record: found
          • Abstract: found
          • Article: not found

          Ribo-gnome: the big world of small RNAs.

          Small RNA guides--microRNAs, small interfering RNAs, and repeat-associated small interfering RNAs, 21 to 30 nucleotides in length--shape diverse cellular pathways, from chromosome architecture to stem cell maintenance. Fifteen years after the discovery of RNA silencing, we are only just beginning to understand the depth and complexity of how these RNAs regulate gene expression and to consider their role in shaping the evolutionary history of higher eukaryotes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Inducible microRNA-155 feedback promotes type I IFN signaling in antiviral innate immunity by targeting suppressor of cytokine signaling 1.

            Effective recognition of viral infection and subsequent triggering of antiviral innate immune responses are essential for the host antiviral defense, which is tightly regulated by multiple regulators, including microRNAs. Our previous study showed that a panel of microRNAs, including miR-155, was markedly upregulated in macrophages upon vesicular stomatitis virus infection; however, the biological function of miR-155 during viral infection remains unknown. In this paper, we show that RNA virus infection induces miR-155 expression in macrophages via TLR/MyD88-independent but retinoic acid-inducible gene I/JNK/NF-κB-dependent pathway. And the inducible miR-155 feedback promotes type I IFN signaling, thus suppressing viral replication. Furthermore, suppressor of cytokine signaling 1 (SOCS1), a canonical negative regulator of type I IFN signaling, is targeted by miR-155 in macrophages, and SOCS1 knockdown mediates the enhancing effect of miR-155 on type I IFN-mediated antiviral response. Therefore, we demonstrate that inducible miR-155 feedback positively regulates host antiviral innate immune response by promoting type I IFN signaling via targeting SOCS1.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Influenza vaccines: challenges and solutions.

              Vaccination is the best method for the prevention and control of influenza. Vaccination can reduce illness and lessen severity of infection. This review focuses on how currently licensed influenza vaccines are generated in the U.S., why the biology of influenza poses vaccine challenges, and vaccine approaches on the horizon that address these challenges.
                Bookmark

                Author and article information

                Contributors
                lin.liu@okstate.edu
                Journal
                Cell Microbiol
                Cell. Microbiol
                10.1111/(ISSN)1462-5822
                CMI
                Cellular Microbiology
                John Wiley and Sons Inc. (Hoboken )
                1462-5814
                1462-5822
                25 January 2019
                May 2019
                : 21
                : 5 ( doiID: 10.1111/cmi.v21.5 )
                : e13001
                Affiliations
                [ 1 ] Oklahoma Center for Respiratory and Infectious Diseases Oklahoma State University Stillwater Oklahoma USA
                [ 2 ] Lundberg‐Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences Oklahoma State University Stillwater Oklahoma USA
                Author notes
                [*] [* ] Correspondence

                Lin Liu, Ph.D., Department of Physiological Sciences, Oklahoma State University, 264 McElroy Hall, Stillwater, OK 74078, USA.

                Email: lin.liu@ 123456okstate.edu

                Author information
                https://orcid.org/0000-0002-4811-4897
                Article
                CMI13001 CMI-18-0054.R2
                10.1111/cmi.13001
                6459727
                30650225
                b2ca2745-1463-4b6e-8e04-c604b5ac854c
                © 2019 John Wiley & Sons Ltd

                This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency.

                History
                : 05 March 2018
                : 12 December 2018
                : 19 December 2018
                Page count
                Figures: 12, Tables: 1, Pages: 20, Words: 10538
                Funding
                Funded by: National Institutes of Health , open-funder-registry 10.13039/100000098;
                Award ID: AI121591
                Award ID: GM103648
                Award ID: HL116876
                Award ID: HL135152
                Funded by: Oklahoma Center for Adult Stem Cell Research
                Funded by: Lundberg‐Kienlen Endowment fund
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                May 2019
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.0 mode:remove_FC converted:16.04.2020

                Microbiology & Virology
                cell cycle arrest,influenza a virus,microrna,mir‐193b,vrnp nuclear import,wnt/β‐catenin signaling

                Comments

                Comment on this article