Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

2′-O-methylation in mRNA disrupts tRNA decoding during translation elongation

Abstract

Chemical modifications of mRNA may regulate many aspects of mRNA processing and protein synthesis. Recently, 2′-O-methylation of nucleotides was identified as a frequent modification in translated regions of human mRNA, showing enrichment in codons for certain amino acids. Here, using single-molecule, bulk kinetics and structural methods, we show that 2′-O-methylation within coding regions of mRNA disrupts key steps in codon reading during cognate tRNA selection. Our results suggest that 2′-O-methylation sterically perturbs interactions of ribosomal-monitoring bases (G530, A1492 and A1493) with cognate codon–anticodon helices, thereby inhibiting downstream GTP hydrolysis by elongation factor Tu (EF-Tu) and A-site tRNA accommodation, leading to excessive rejection of cognate aminoacylated tRNAs in initial selection and proofreading. Our current and prior findings highlight how chemical modifications of mRNA tune the dynamics of protein synthesis at different steps of translation elongation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Multiple cognate tRNAs are rejected during decoding of the 2′-O-methylated codon.
Fig. 2: Codon context alters the magnitude of a 2′-O-methylation-induced stall.
Fig. 3: 2′-O-methylation prolongs time between the initial tRNA binding and its full accommodation event.
Fig. 4: Bulk kinetics measurements of (kcat/Km)GTP with modified (AAmA) and unmodified (AAA) codons at 5 mM Mg2+.
Fig. 5: The effect of 2′-O-methylation on proofreading.
Fig. 6: The delay in GTP hydrolysis by 2′-O-methylation is due to the inactivation of the monitoring bases.
Fig. 7: Mechanism of epitranscriptomic-induced translation-elongation stall.

Similar content being viewed by others

References

  1. Zhao, B. S., Roundtree, I. A. & He, C. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 18, 31–42 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Liu, N. et al. N 6-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature 518, 560–564 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Fu, Y., Dominissini, D., Rechavi, G. & He, C. Gene expression regulation mediated through reversible m6A RNA methylation. Nat. Rev. Genet. 15, 293–306 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Choi, J. et al. N 6-methyladenosine in mRNA disrupts tRNA selection and translation-elongation dynamics. Nat. Struct. Mol. Biol. 23, 110–115 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dai, Q. et al. Nm-seq maps 2′-O-methylation sites in human mRNA with base precision. Nat. Methods 14, 695–698 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Decatur, W. A. & Fournier, M. J. rRNA modifications and ribosome function. Trends Biochem. Sci. 27, 344–351 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Somme, J. et al. Characterization of two homologous 2′-O-methyltransferases showing different specificities for their tRNA substrates. RNA 20, 1257–1271 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cavaillé, J., Nicoloso, M. & Bachellerie, J.-P. Targeted ribose methylation of RNA in vivo directed by tailored antisense RNA guides. Nature 383, 732–735 (1996).

    Article  PubMed  Google Scholar 

  9. Kiss-László, Z., Henry, Y., Bachellerie, J. P., Caizergues-Ferrer, M. & Kiss, T. Site-specific ribose methylation of preribosomal RNA: a novel function for small nucleolar RNAs. Cell 85, 1077–1088 (1996).

    Article  PubMed  Google Scholar 

  10. Shubina, M. Y., Musinova, Y. R. & Sheval, E. V. Nucleolar methyltransferase fibrillarin: evolution of structure and functions. Biochemistry 81, 941–950 (2016).

    CAS  PubMed  Google Scholar 

  11. Cummins, L. L. et al. Characterization of fully 2′-modified oligoribonucleotide hetero- and homoduplex hybridization and nuclease sensitivity. Nucleic Acids Res. 23, 2019–2024 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Majlessi, M., Nelson, N. C. & Becker, M. M. Advantages of 2′-O-methyl oligoribonucleotide probes for detecting RNA targets. Nucleic Acids Res. 26, 2224–2229 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hoernes, T. P. et al. Nucleotide modifications within bacterial messenger RNAs regulate their translation and are able to rewire the genetic code. Nucleic Acids Res. 44, 852–862 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Yoshizawa, S., Fourmy, D. & Puglisi, J. D. Recognition of the codon-anticodon helix by ribosomal RNA. Science 285, 1722–1725 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Ogle, J. M. et al. Recognition of cognate transfer RNA by the 30S ribosomal subunit. Science 292, 897–902 (2001).

  16. Uemura, S. et al. Real-time tRNA transit on single translating ribosomes at codon resolution. Nature 464, 1012–1017 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chen, J. et al. High-throughput platform for real-time monitoring of biological processes by multicolor single-molecule fluorescence. Proc. Natl. Acad. Sci. USA 111, 664–669 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Dorywalska, M. et al. Site-specific labeling of the ribosome for single-molecule spectroscopy. Nucleic Acids Res. 33, 182–189 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Aitken, C. E. & Puglisi, J. D. Following the intersubunit conformation of the ribosome during translation in real time. Nat. Struct. Mol. Biol. 17, 793–800 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chen, J., Tsai, A., Petrov, A. & Puglisi, J. D. Nonfluorescent quenchers to correlate single-molecule conformational and compositional dynamics. J. Am. Chem. Soc. 134, 5734–5737 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kurland, C. G. & Ehrenberg, M. Optimization of translation accuracy. Prog. Nucleic Acid Res. Mol. Biol. 31, 191–219 (1984).

    Article  CAS  PubMed  Google Scholar 

  22. Blanchard, S. C. Jr, Gonzalez, R. L., Kim, H. D., Chu, S. & Puglisi, J. D. tRNA selection and kinetic proofreading in translation. Nat. Struct. Mol. Biol. 11, 1008–1014 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Loveland, A. B., Demo, G., Grigorieff, N. & Korostelev, A. A. Ensemble cryo-EM elucidates the mechanism of translation fidelity. Nature 546, 113–117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ninio, J. Kinetic amplification of enzyme discrimination. Biochimie 57, 587–595 (1975).

    Article  CAS  PubMed  Google Scholar 

  25. Hopfield, J. J. Kinetic proofreading: a new mechanism for reducing errors in biosynthetic processes requiring high specificity. Proc. Natl. Acad. Sci. USA 71, 4135–4139 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ogle, J. M., Murphy, F. V., Tarry, M. J. & Ramakrishnan, V. Selection of tRNA by the ribosome requires a transition from an open to a closed form. Cell 111, 721–732 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Satpati, P., Sund, J. & Åqvist, J. Structure-based energetics of mRNA decoding on the ribosome. Biochemistry 53, 1714–1722 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Vendeix, F. A. et al. Human tRNA(Lys3)(UUU) is pre-structured by natural modifications for cognate and wobble codon binding through keto-enol tautomerism. J. Mol. Biol. 416, 467–485 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Demirci, H. et al. Modification of 16 S ribosomal RNA by the KsgA methyltransferase restructures the 30S subunit to optimize ribosome function. RNA 16, 2319–2324 (2010).

  30. Wimberly, B. T. et al. Structure of the 30S ribosomal subunit. Nature 407, 327–339 (2000).

  31. Zhou, H. et al. m1A and m1G disrupt A-RNA structure through the intrinsic instability of Hoogsteen base pairs. Nat. Struct. Mol. Biol. 23, 803–810 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Roost, C. et al. Structure and thermodynamics of N6-methyladenosine in RNA: a spring-loaded base modification. J. Am. Chem. Soc. 137, 2107–2115 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ieong, K.-W., Uzun, Ü., Selmer, M. & Ehrenberg, M. Two proofreading steps amplify the accuracy of genetic code translation. Proc. Natl. Acad. Sci. USA 113, 13744–13749 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Richter, J. D. & Coller, J. Pausing on polyribosomes: make way for elongation in translational control. Cell 163, 292–300 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Marshall, R. A., Dorywalska, M. & Puglisi, J. D. Irreversible chemical steps control intersubunit dynamics during translation. Proc. Natl. Acad. Sci. USA 105, 15364–15369 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, J., Petrov, A., Tsai, A., O’Leary, S. E. & Puglisi, J. D. Coordinated conformational and compositional dynamics drive ribosome translocation. Nat. Struct. Mol. Biol. 20, 718–727 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wold, F. & Ballou, C. E. Studies on the enzyme enolase. I. Equilibrium studies. J. Biol. Chem. 227, 301–312 (1957).

    CAS  PubMed  Google Scholar 

  38. Johansson, M., Zhang, J. & Ehrenberg, M. Genetic code translation displays a linear trade-off between efficiency and accuracy of tRNA selection. Proc. Natl. Acad. Sci. USA 109, 131–136 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Zhang, J., Ieong, K.-W., Johansson, M. & Ehrenberg, M. Accuracy of initial codon selection by aminoacyl-tRNAs on the mRNA-programmed bacterial ribosome. Proc. Natl. Acad. Sci. USA 112, 9602–9607 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kabsch, W. Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, (213–221 (2010).

    Google Scholar 

  42. Kurata, S. et al. Modified uridines with C5-methylene substituents at the first position of the tRNA anticodon stabilize U.G wobble pairing during decoding. J. Biol. Chem. 283, 18801–18811 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. The PyMOL Molecular Graphics System v.1.7.4 (Schrödinger, LLC, New York, NY, 2015).

Download references

Acknowledgements

This work was supported by the US National Institutes of Health (NIH) grants GM51266 and GM113078 to J.D.P. and by grants from the Knut and Alice Wallenberg Foundation (RiboCORE), the Swedish Research Council and the Human Frontier Science Program to M.E., the Kahn Family Foundation to D.D. and G.R., the Ernest and Bonnie Beutler Research Program, Flight Attendant Medical Research Institute (FAMRI) and the Israeli Centers of Excellence (I-CORE) Program (ISF grants no. 41/11 and no. 1796/12) to G.R., a Human Frontier Science Program long-term fellowship to D.D., by the NSF Science and Technology Centers grant NSF-1231306 (Biology with X-ray Lasers, BioXFEL) to H.D., by a Stanford Bio-X fellowship to J.C. and A. Prabhakar and by a Knut and Alice Wallenberg Foundation postdoc fellowship to J.W. Portions of this research were carried out at the Stanford Synchrotron Radiation Lightsource (SSRL), a national user facility operated by Stanford University on behalf of the US Department of Energy, US Office of Basic Energy Sciences. The SSRL Structural Molecular Biology Program is supported by the US Department of Energy, Office of Biological and Environmental Research, NIH, US National Center for Research Resources, Biomedical Technology Program, and the US National Institute of General Medical Sciences. C.H. is supported by the Howard Hughes Medical Institute (HHMI). We thank P. Agris (University of Albany) for a human ASL reagent and members of Puglisi laboratory for discussion.

Author information

Authors and Affiliations

Authors

Contributions

J.C., G.I., H.D. and K.-W.I. performed all experiments and data analysis; J.C. performed single-molecule experiments, with the help of J.W., A. Prabhakar and A. Petrov in material preparation; G.I. and K.-W.I. performed bulk kinetics experiments; H.D. performed X-ray crystallography. D.D., G.R. and C.H. provided reagents and conceived the project with J.C., H.D., K.-W.I., M.E. and J.D.P. J.C., G.I., H.D., J.W., M.E. and J.D.P. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Joseph D. Puglisi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 tRNA-tRNA FRET reveals the tRNA sampling state is A*/T-site-bound state.

(a) A representative trace from tRNA-tRNA FRET experiments. On the modified codon (AAmA), we observe tRNA is bound to the A*/T site during sampling (prior to GTP-hydrolysis activation; FRET efficiency near 0.5), followed by the accommodation event (FRET efficiency near 0.7).(b) Contour plots of FRET-efficiency trajectories, generated by post-synchronizing traces to the successful tRNA accommodation events on AAmA codon at 15 mM Mg2+ concentration (n = 369). (c) Contour plots of FRET-efficiency trajectories, generated by post-synchronizing traces to the successful tRNA accommodation events on AAA codon at 15 mM Mg2+ concentration (n = 371). (d) Contour plots of FRET-efficiency trajectories, generated by post-synchronizing traces to the tRNA sampling events on AAmA codon at 15 mM Mg2+ concentration (n = 1244). (e) Contour plots of FRET-efficiency trajectories, generated by post-synchronizing traces to the tRNA sampling events on AAmA codon at 15 mM Mg2+ concentration in the presence of GDPNP (n = 565). (f)Contour plots of FRET-efficiency trajectories, generated by post-synchronizing traces to the tRNA sampling events on AAA codon at 15 mM Mg2+ concentration in the presence of GDPNP (n = 420). (g) tRNA-tRNA FRET efficiencies assigned to different stages of tRNA selection: Upon the initial binding of tRNA in the form of TC, the codon-anticodon interaction stabilizes tRNA to the A*/T-site (FRET-efficiency near 0.5), where monitoring bases has not made contact with codon-anticodon helices to activate GTP-hydrolysis. Activation of monitoring bases slightly moves tRNA to the A/T-site, which facilitates GTP-hydrolysis (FRET-efficiency near 0.6). GTP-hydrolysis followed by the peptide-bond formation accommodates tRNA to the A site (FRET-efficiency near 0.7).

Supplementary Figure 2 fMet consumption over time with Lys-tRNA only or with Lys-tRNA depleted bulk aa-tRNA.

0.2 µM ribosomes initiated with [3H]fMet-tRNAfMet and AAA or AAmA codon in the A site were reacted to 2 µM EF-Tu and 100 µM total E. coli tRNA charged with all 19 amino acids except lysine or only lysine, as indicated. The experiment was performed in polymix buffer containing 15 mM total Mg2+. The decrease of the fMet-tRNAfMet fraction over time reveals a corresponding increase in peptide bond formation. In the presence of Lys-tRNALys and unmodified AAA codon all available fMet was consumed by fMet-Lys formation (filled-circle data points), but in the absence of Lys-tRNALys and presence of charged bulk-tRNA the extent of peptide bond formation was negligible both for the modified AAmA (filled-triangle data points) and unmodified AAA (filled-diamond data points) codon.

Supplementary Figure 3 Aminoglycosides decrease the time lag between tRNA binding and accommodation.

Contour plots of the normalized Cy3B and Cy5 intensity trajectories, generated by post-synchronizing to the ribosomal intersubunit rotation event upon the peptidyl transfer reaction. Without any drug (lower-left panel), the Cy5 signal (upper plots; binding of (Cy5)-tRNALys to the ribosome) happens about 2 seconds earlier than the unquenching of Cy3B signal (lower plots; intersubunit rotation to the rotated state after peptidyl-transfer reaction) on the 2′-O-methylated codon. This time delay (indicated as Rotation Lag with a red bar) is minimized in the presence of paromomycin or neomycin (middle and right panels).

Supplementary Figure 4 X-ray crystal structures without paromomycin.

Structures of the decoding center (ASL and A1492, A1493 and G530 monitoring bases) while decoding (a) the unmodified, (b) the first base modified, (c) the second base modified, and (d) the third base modified Lys codon, soaked in the 30S crystals in the absence of paromomycin. Without paromomycin, crystal samples prepared in parallel with the modified Lys codon did not exhibit successful binding of ASL and mRNA nucleotides to the ribosome, marked by the lack of corresponding 2Fo-Fc electron densities contoured at 1.5 sigma level.

Supplementary Figure 5 Decoding center composite omit maps without paromomycin.

Composite omit 2mFo − DFc electron density maps of decoding center region (ASL and A1492, A1493 and G530 monitoring bases) while decoding (a) the unmodified, (b) the first base modified, (c) the second base modified, and (d) the third base modified Lys codon, soaked in the 30S crystals in the absence of paromomycin. Without paromomycin, crystal samples prepared in parallel with the modified Lys codon did not exhibit successful binding of ASL and mRNA nucleotides to the ribosome, marked by the lack of corresponding 2Fo-DFc composite omit map electron densities.

Supplementary Figure 6 Simple FoFc omit maps of unmodified and modified mRNA bases.

X-ray crystallography structures of the decoding center (UUU anticodon and A1492, A1493 and G530 monitoring bases) while decoding unmodified (AAA) (a) the first base unmodified (b) the second base unmodified (c) the third base unmodified (d) the first base modified (AmAA), (e) the second base modified (AAmA), and (f) the third base modified Lys codon (AAAm), soaked in the 30S subunit crystals in the presence of paromomycin.

Supplementary Figure 7 X-ray crystal structures soaked with paromomycin.

Structures of the decoding center (ASL and A1492, A1493 and G530 monitoring bases) while decoding (a) the unmodified, (b) the first base modified, (c) the second base modified, and (d) the third base modified Lys codon, soaked in the 30S crystals in the presence of paromomycin. 2Fo-Fc electron density maps are contoured at 1.5 sigma level.

Supplementary Figure 8 Mechanistic model of 2′-O-methylation affecting tRNA decoding.

(a) The 2′-O-methylation within the A-site codon sterically disrupts the activation of monitoring bases (A1492 shown as it interacts with the second base within the A site codon). (b) Schematics of tRNA selection to the 2′-O-methylated codon: While the 2′-O-methylation disrupts the activation of monitoring bases, aminoglycosides such as paromomycin and neomycin counter its effect by forcing the activation of monitoring bases. The disruption of interactions among monitoring bases and mRNA-tRNA duplex may also delay EF-Tu•GDP dissociation, favoring tRNA rejection by aa-tRNA•EF-Tu•GDP dissociation. The increase of Mg2+ concentration may slow down both aa-tRNA•EF-Tu•GTP and aa-tRNA•EF-Tu•GDP dissociation rate, favoring tRNA accommodation on the 2′-O-methylated codon during the initial selection process and proofreading steps.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Notes 1–3

Life Sciences Reporting Summary

Supplementary Dataset 1

Lifetimes and ribosome survival plots within the coding region for Figure 1.

Supplementary Dataset 2

Lifetimes and ribosome survival plots within the coding region for Figure 2.

Supplementary Dataset 3

Source data and statistical measures underlying figures.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Choi, J., Indrisiunaite, G., DeMirci, H. et al. 2′-O-methylation in mRNA disrupts tRNA decoding during translation elongation. Nat Struct Mol Biol 25, 208–216 (2018). https://doi.org/10.1038/s41594-018-0030-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0030-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing