Here we seek to understand the role of RNA interference (RNAi) machinery in mammalian antiviral defense. RNAi is a nucleic acid sequence-specific means of gene silencing guided by short, double-stranded RNA. Plants, insects and C. elegans use RNAi to combat viral infections, however mammals evolved with alternative antiviral mechanisms, including pattern recognition receptors (PRRs) and components of the inflammasome, leading to the production of type I IFN and other cytokines to achieve an antiviral state. The key RNAi effector proteins, Dicer and Argonautes (AGO)1-4 are functional in mammalian cells, being essential for development and regulation of the microRNA and RNAi systems. However, any role for RNAi machinery in mammalian host- virus interactions remains to be fully characterized. In addition, whether redundancy between the four mammalian AGOs exits, for executing RNAi or other small RNA-mediated repression in any physiological context remains unclear. This work is significant because this ancient mechanism of innate immunity may be important for defense against many human viruses. To study the role of RNAi machinery in mammalian immunology, we will use mice lacking individual AGO proteins, which will serve as primary tools for the proposed studies. In preliminary results reported here, we demonstrate profound but differential contributions of AGOs 2 and 4 to mammalian antiviral responses. Antiviral responses are markedly dimished in the absence of AGO2 catalytic activity or AGO4 protein. Moreover, AGO1 and 3 appear to play minimal roles in antiviral defense. However, a lack of AGO2 protein resulted in reduced virus levels, suggesting AGO2 negatively regulates other antiviral pathways. This application aims to comprehensively dissect the functions of individual Argonautes in the antiviral response. Specifically, the studies proposed in aim 1 will characterize the role of AGO2, the only argonaute with slicing capacity, in silencing virus.
Aim 2 proposes to decipher how AGO4 contributes to the mammalian antiviral responses.
Both aims will determine whether AGOs act autonomously or how they aid or compete with existing PRR and type I IFN mammalian antiviral defense mechanisms.
Aim 3 will assess what host and virus-derived RNA populations bind to AGOs 2 and 4 in influenza A infected macrophages. These virus-derived RNAs will then be tested for their virus silencing capacity. This work will elucidate the undefined roles of individual AGO proteins in antiviral defense and may offer new therapeutic targets for the treatment of viral infections.

Public Health Relevance

RNA interference (RNAi) is a highly conserved gene regulation process that plants, flies and worms use to directly attack viruses. Humans possess all of the tools for RNAi and use it to regulate genes, but whether it is used for antiviral defens is unknown. This study aims to uncover this immune process which will open new avenues to treat virus infections, such as influenza and HIV.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI107087-05
Application #
9506676
Study Section
Immunity and Host Defense (IHD)
Program Officer
Jiang, Chao
Project Start
2014-07-01
Project End
2019-06-30
Budget Start
2018-07-01
Budget End
2019-06-30
Support Year
5
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Massachusetts General Hospital
Department
Type
DUNS #
073130411
City
Boston
State
MA
Country
United States
Zip Code
Jeffrey, Kate L; Li, Yang; Ding, Shou-Wei (2017) Reply to 'Questioning antiviral RNAi in mammals'. Nat Microbiol 2:17053
Fusco, Dahlene N; Pratt, Henry; Kandilas, Stephen et al. (2017) HELZ2 Is an IFN Effector Mediating Suppression of Dengue Virus. Front Microbiol 8:240
Murano, Tatsuro; Najibi, Mehran; Paulus, Geraldine L C et al. (2017) Transcription factor TFEB cell-autonomously modulates susceptibility to intestinal epithelial cell injury in vivo. Sci Rep 7:13938
Mehta, Stuti; Cronkite, D Alexander; Basavappa, Megha et al. (2017) Maintenance of macrophage transcriptional programs and intestinal homeostasis by epigenetic reader SP140. Sci Immunol 2:
Li, Yang; Basavappa, Megha; Lu, Jinfeng et al. (2016) Induction and suppression of antiviral RNA interference by influenza A virus in mammalian cells. Nat Microbiol 2:16250
Mehta, Stuti; Jeffrey, Kate L (2015) Beyond receptors and signaling: epigenetic factors in the regulation of innate immunity. Immunol Cell Biol 93:233-44