Immune Netw.  2016 Aug;16(4):249-255. 10.4110/in.2016.16.4.249.

Dependence of RIG-I Nucleic Acid-Binding and ATP Hydrolysis on Activation of Type I Interferon Response

Affiliations
  • 1Department of Bioscience and Biotechnology, Konkuk University, Seoul 05902, Korea. kimde@konkuk.ac.kr

Abstract

Exogenous nucleic acids induce an innate immune response in mammalian host cells through activation of the retinoic acid-inducible gene I (RIG-I). We evaluated RIG-I protein for RNA binding and ATPase stimulation with RNA ligands to investigate the correlation with the extent of immune response through RIG-I activation in cells. RIG-I protein favored blunt-ended, double-stranded RNA (dsRNA) ligands over sticky-ended dsRNA. Moreover, the presence of the 5'-triphosphate (5'-ppp) moiety in dsRNA further enhanced binding affinity to RIG-I. Two structural motifs in RNA, blunt ends in dsRNA and 5'-ppp, stimulated the ATP hydrolysis activity of RIG-I. These structural motifs also strongly induced IFN expression as an innate immune response in cells. Therefore, we suggest that IFN induction through RIG-I activation is mainly determined by structural motifs in dsRNA that increase its affinity for RIG-I protein and stimulate ATPase activity in RIG-I.

Keyword

Retinoic acid inducible gene I (RIG-I); Interferon induction; 5'-terminal triphosphate; ATP hydrolysis; RNA binding

MeSH Terms

Adenosine Triphosphatases
Adenosine Triphosphate*
Hydrolysis*
Immunity, Innate
Interferon Type I*
Ligands
Nucleic Acids
RNA
RNA, Double-Stranded
Adenosine Triphosphatases
Adenosine Triphosphate
Interferon Type I
Ligands
Nucleic Acids
RNA
RNA, Double-Stranded

Figure

  • Figure 1 Binding affinities of various types of duplex nucleic acid ligands to RIG-I protein. (A) Increasing amounts of RIG-I protein were incubated with each duplex nucleic acid ligand; the dsRNA ligands included 3'-end overhang (47R/25RI), 5'-end overhang (47R/25RII), double overhang (47R/25RIII)' and RNA:DNA hybrids including 3'-end overhang (47R/25DI) and 5'-end overhang (47R/25DII). Nucleic acid ligands complexed with the RIG-I protein appeared as slowly migrating bands on 2% agarose gel after the electrophoretic mobility shift assay (EMSA). The nucleic acid band intensities were quantitated by ImageJ software, and the extent of nucleic acid ligands bound to the RIG-I protein were plotted as normalized values relative to the control (No RIG-I protein). Each curve was fitted to the Hill equation; Y=A×Lh/(Bh+Lh), where Y represents for relative fraction of RNA ligands bound to RIG-I at each RNA ligand concentration (L), A represents for the amount of RNA ligands complexed to RIG-I at equilibrium, and B and h represents apparent dissociation constant (i.e. Kd) and the Hill coefficient, respectively. Each apparent Kd (in µM) value was obtained from the fitting of the curve to the above equation using Sigma Plot, and represented as the bar graph (inset). (B) Increasing amounts of RIG-I protein were incubated with double-stranded nucleic acids with 3'-end overhang and 5'-end triphosphate (3P-47R/25RI and 3P-47R/25DI) or 5'-end-OH (47R/25RI and 47R/25DI). After the EMSA, the migration of the nucleic acid ligands was visualized on the gel. The upper band represents nucleic acids bound to RIG-I, whereas the bottom band shows unbound free nucleic acid ligands. The nucleic acid band intensities were quantitated by ImageJ software, and the extent of nucleic acid ligands bound to the RIG-I protein were plotted as normalized values relative to the control (No RIG-I protein). Each curve was fitted to the Hill equation; the amplitudes of each curve were different at equilibrium.

  • Figure 2 Stimulation of ATP hydrolysis by RIG-I with various types of nucleic acid ligands. ATP (200 M) was mixed with RIG-I (20 nM) in the presence of each nucleic acid ligand (0.5 nM). (A) ATP hydrolysis by RIG-I was monitored over time in the presence of dsRNAs including 3'-end overhang (47R/25RI), 5'-end overhang (47R/25RII), and double overhang (47R/25RIII). The thin-layer chromatography plates show the progress of the ATP hydrolysis reaction, in which ATP hydrolysis products (radioactive inorganic phosphate, Pi*) and radioactive substrates ATP* were resolved and quantified. (B) The ATPase activity of RIG-I over time was monitored in the presence of ssRNAs including 5'-ppp RNA (3P-47R) and ssRNA or ssDNA with a 5'-end hydroxyl group (47R, 25RI, and 25DI).

  • Figure 3 Interferon induction in lung epithelial cells with various types of nucleic acid ligands through RIG-I activation. A549 cells were transfected with the 5'-ppp or 5'-HO RNA substrates or the RNA:DNA hybrid substrate. A549 cells containing the pGL3-IFNβ reporter gene were transfected with each duplex RNA ligand at two different concentrations (10 nM or 50 nM). The luciferase reporter assay was conducted at 18 h post-transfection with each RNA ligand. Activation of the IFN-β promoter with the exogenous RNA ligand is shown in the graph as the fold-index relative to a mock transfection control (transfection reagent only). Transfections with yeast tRNA and poly I:C were performed for negative and positive controls, respectively. The unit fold was set to the IFN expression observed in the mock transfection. The values shown are the means±S.D. of triplicate experiments.


Reference

1. Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol. 2015; 33:257–290.
Article
2. Wu J, Chen ZJ. Innate immune sensing and signaling of cytosolic nucleic acids. Annu Rev Immunol. 2014; 32:461–488.
Article
3. Thompson AJ, Locarnini SA. Toll-like receptors, RIG-I-like RNA helicases and the antiviral innate immune response. Immunol Cell Biol. 2007; 85:435–445.
Article
4. Gee P, Chua PK, Gevorkyan J, Klumpp K, Najera I, Swinney DC, Deval J. Essential role of the N-terminal domain in the regulation of RIG-I ATPase activity. J Biol Chem. 2008; 283:9488–9496.
Article
5. O'Neill LA, Bowie AG. The powerstroke and camshaft of the RIG-I antiviral RNA detection machine. Cell. 2011; 147:259–261.
6. Jiang F, Ramanathan A, Miller MT, Tang GQ, Gale M Jr, Patel SS, Marcotrigiano J. Structural basis of RNA recognition and activation by innate immune receptor RIG-I. Nature. 2011; 479:423–427.
Article
7. Baek SE, Kim H, Kim KB, Yoon S, Choe J, Suh W, Jeong YJ, Cho YH, Kim DE. Dual effects of duplex RNA harboring 5'-terminal triphosphate on gene silencing and RIG-I mediated innate immune response. Biochem Biophys Res Commun. 2015; 456:591–597.
Article
8. Ranjith-Kumar CT, Murali A, Dong W, Srisathiyanarayanan D, Vaughan R, Ortiz-Alacantara J, Bhardwaj K, Li X, Li P, Kao CC. Agonist and antagonist recognition by RIG-I, a cytoplasmic innate immunity receptor. J Biol Chem. 2009; 284:1155–1165.
Article
9. Wang Y, Ludwig J, Schuberth C, Goldeck M, Schlee M, Li H, Juranek S, Sheng G, Micura R, Tuschl T, Hartmann G, Patel DJ. Structural and functional insights into 5'-ppp RNA pattern recognition by the innate immune receptor RIG-I. Nat Struct Mol Biol. 2010; 17:781–787.
Article
10. Kowalinski E, Lunardi T, McCarthy AA, Louber J, Brunel J, Grigorov B, Gerlier D, Cusack S. Structural basis for the activation of innate immune pattern-recognition receptor RIG-I by viral RNA. Cell. 2011; 147:423–435.
Article
11. Gack MU, Shin YC, Joo CH, Urano T, Liang C, Sun L, Takeuchi O, Akira S, Chen Z, Inoue S, Jung JU. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature. 2007; 446:916–920.
Article
12. Wu B, Peisley A, Tetrault D, Li Z, Egelman EH, Magor KE, Walz T, Penczek PA, Hur S. Molecular imprinting as a signal-activation mechanism of the viral RNA sensor RIG-I. Mol Cell. 2014; 55:511–523.
Article
13. Foy E, Li K, Wang C, Sumpter R Jr, Ikeda M, Lemon SM, Gale M Jr. Regulation of interferon regulatory factor-3 by the hepatitis C virus serine protease. Science. 2003; 300:1145–1148.
Article
14. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004; 5:730–737.
Article
15. Myong S, Cui S, Cornish PV, Kirchhofer A, Gack MU, Jung JU, Hopfner KP, Ha T. Cytosolic viral sensor RIG-I is a 5'-triphosphate-dependent translocase on double-stranded RNA. Science. 2009; 323:1070–1074.
Article
16. Lassig C, Matheisl S, Sparrer KM, de Oliveira Mann CC, Moldt M, Patel JR, Goldeck M, Hartmann G, Garcia-Sastre A, Hornung V, Conzelmann KK, Beckmann R, Hopfner KP. ATP hydrolysis by the viral RNA sensor RIG-I prevents unintentional recognition of self-RNA. Elife. 2015; 4:e10859.
17. Lee SY, Jung HY, Kim TO, Im DW, You KY, Back JM, Kim Y, Kim HJ, Shin W, Heo YS. Cloning, purification, crystallization and preliminary X-ray crystallographic analysis of the N-terminal domain of DEAD-box RNA helicase from Staphylococcus aureus strain Mu50. Acta Crystallogr Sect F Struct Biol Cryst Commun. 2010; 66:1674–1676.
Article
18. Lee B, Kim KB, Oh S, Choi JS, Park JS, Min DH, Kim DE. Suppression of hepatitis C virus genome replication in cells with RNA-cleaving DNA enzymes and short-hairpin RNA. Oligonucleotides. 2010; 20:285–296.
Article
19. Zhu FX, King SM, Smith EJ, Levy DE, Yuan Y. A Kaposi's sarcoma-associated herpesviral protein inhibits virus-mediated induction of type I interferon by blocking IRF-7 phosphorylation and nuclear accumulation. Proc Natl Acad Sci U S A. 2002; 99:5573–5578.
Article
20. Lee MK, Kim HE, Park EB, Lee J, Kim KH, Lim K, Yum S, Lee YH, Kang SJ, Lee JH, Choi BS. Structural features of influenza A virus panhandle RNA enabling the activation of RIG-I independently of 5'-triphosphate. Nucleic Acids Res. 2016; DOI: 10.1093/nar/gkw525.
Article
Full Text Links
  • IN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr