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:

Structural basis of amino acid surveillance by higher-order tRNA-mRNA interactions

Abstract

Amino acid availability in Gram-positive bacteria is monitored by T-box riboswitches. T-boxes directly bind tRNAs, assess their aminoacylation state, and regulate the transcription or translation of downstream genes to maintain nutritional homeostasis. Here, we report cocrystal and cryo-EM structures of Geobacillus kaustophilus and Bacillus subtilis T-box–tRNA complexes, detailing their multivalent, exquisitely selective interactions. The T-box forms a U-shaped molecular vise that clamps the tRNA, captures its 3′ end using an elaborate ‘discriminator’ structure, and interrogates its aminoacylation state using a steric filter fashioned from a wobble base pair. In the absence of aminoacylation, T-boxes clutch tRNAs and form a continuously stacked central spine, permitting transcriptional readthrough or translation initiation. A modeled aminoacyl disrupts tRNA-T-box stacking, severing the central spine and blocking gene expression. Our data establish a universal mechanism of amino acid sensing on tRNAs and gene regulation by T-box riboswitches and exemplify how higher-order RNA-RNA interactions achieve multivalency and specificity.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Overall structure of the T-box discriminator–tRNA complex.
Fig. 2: The A-minor latch and pseudohelix stabilize tRNA-T-box interactions.
Fig. 3: Structural basis of tRNA aminoacylation sensing by the T-box discriminator.
Fig. 4: Docking of stem III with helix A2 reinforces and rigidifies complex structure.
Fig. 5: 4.9-Å resolution cryo-EM map and structure of a full-length B. subtilis glyQS T-box-tRNAGly complex.
Fig. 6: The T-box central spine and structural comparisons.
Fig. 7: Mechanistic model of a cotranscriptionally acting T-box riboswitch.

Similar content being viewed by others

Data availability

Atomic coordinates and structure factor amplitudes for the T-box discriminator in complex with tRNAGly have been deposited at the Protein Data Bank (PDB) under accession code PDB 6PMO. Cryo-EM structure of the full-length T-box–tRNA complex and map have been deposited to Electron Microscopy Data Bank under EMD-20416 and PDB 6POM. All other data are available upon reasonable request.

References

  1. Chantranupong, L., Wolfson, R. L. & Sabatini, D. M. Nutrient-sensing mechanisms across evolution. Cell 161, 67–83 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Saxton, R. A. et al. Structural basis for leucine sensing by the Sestrin2-mTORC1 pathway. Science 351, 53–58 (2016).

    CAS  PubMed  Google Scholar 

  3. Chantranupong, L. et al. The CASTOR proteins are arginine sensors for the mTORC1 Pathway. Cell 165, 153–164 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Antson, A. A. et al. Structure of the trp RNA-binding attenuation protein, TRAP, bound to RNA. Nature 401, 235–242 (1999).

    CAS  PubMed  Google Scholar 

  5. Serganov, A. & Patel, D. J. Amino acid recognition and gene regulation by riboswitches. Biochim. Biophys. Acta 1789, 592–611 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Roth, A. & Breaker, R. R. The structural and functional diversity of metabolite-binding riboswitches. Annu. Rev. Biochem. 78, 305–334 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Pang, Y. L., Poruri, K. & Martinis, S. A. tRNA synthetase: tRNA aminoacylation and beyond. Wiley Interdiscip. Rev. RNA 5, 461–480 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Bullwinkle, T. J. & Ibba, M. Emergence and evolution. Top Curr. Chem. 344, 43–87 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Beuning, P. J. & Musier-Forsyth, K. Transfer RNA recognition by aminoacyl-tRNA synthetases. Biopolymers 52, 1–28 (1999).

    CAS  PubMed  Google Scholar 

  10. Yanofsky, C. Attenuation in the control of expression of bacterial operons. Nature 289, 751–758 (1981).

    CAS  PubMed  Google Scholar 

  11. Loveland, A. B. et al. Ribosome*RelA structures reveal the mechanism of stringent response activation. eLife 5, e17029 (2016).

    PubMed  PubMed Central  Google Scholar 

  12. Brown, A., Fernandez, I. S., Gordiyenko, Y. & Ramakrishnan, V. Ribosome-dependent activation of stringent control. Nature 534, 277–280 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Dong, J., Qiu, H., Garcia-Barrio, M., Anderson, J. & Hinnebusch, A. G. Uncharged tRNA activates GCN2 by displacing the protein kinase moiety from a bipartite tRNA-binding domain. Mol. Cell 6, 269–279 (2000).

    CAS  PubMed  Google Scholar 

  14. Zhang, J. & Ferré-D’Amaré, A. R. Direct evaluation of tRNA aminoacylation status by the T-box riboswitch using tRNA-mRNA stacking and steric readout. Mol. Cell 55, 148–155 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Vitreschak, A. G., Mironov, A. A., Lyubetsky, V. A. & Gelfand, M. S. Comparative genomic analysis of T-box regulatory systems in bacteria. RNA 14, 717–735 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gutierrez-Preciado, A., Henkin, T. M., Grundy, F. J., Yanofsky, C. & Merino, E. Biochemical features and functional implications of the RNA-based T-box regulatory mechanism. Microbiol. Mol. Biol. Rev. 73, 36–61 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Saad, N. Y. et al. Two-codon T-box riboswitch binding two tRNAs. Proc. Natl Acad. Sci USA 110, 12756–12761 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Apostolidi, M. et al. A glyS T-box riboswitch with species-specific structural features responding to both proteinogenic and nonproteinogenic tRNAGly isoacceptors. RNA 21, 1790–1806 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Suddala, K. C. & Zhang, J. An evolving tale of two interacting RNAs-themes and variations of the T-box riboswitch mechanism. IUBMB Life 71, 1167–1180 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Frohlich, K. M. et al. Discovery of small-molecule antibiotics against a unique tRNA-mediated regulation of transcription in gram-positive bacteria. ChemMedChem. 14, 758–769 (2019).

    CAS  PubMed  Google Scholar 

  21. Lehmann, J., Jossinet, F. & Gautheret, D. A universal RNA structural motif docking the elbow of tRNA in the ribosome, RNase P and T-box leaders. Nucleic Acids Res. 41, 5494–5502 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, J. & Ferré-D’Amaré, A. R. Co-crystal structure of a T-box riboswitch stem I domain in complex with its cognate tRNA. Nature 500, 363–366 (2013).

    CAS  PubMed  Google Scholar 

  23. Grigg, J. C. & Ke, A. Structural determinants for geometry and information decoding of tRNA by T box leader RNA. Structure 21, 2025–2032 (2013).

    CAS  PubMed  Google Scholar 

  24. Grundy, F. J. & Henkin, T. M. tRNA as a positive regulator of transcription antitermination in B. subtilis. Cell 74, 475–482 (1993).

    CAS  PubMed  Google Scholar 

  25. Grundy, F. J., Winkler, W. C. & Henkin, T. M. tRNA-mediated transcription antitermination in vitro: codon-anticodon pairing independent of the ribosome. Proc. Natl Acad. Sci. USA 99, 11121–11126 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Sherwood, A. V., Frandsen, J. K., Grundy, F. J. & Henkin, T. M. New tRNA contacts facilitate ligand binding in a Mycobacterium smegmatis T box riboswitch. Proc. Natl Acad. Sci. USA 115, 3894–3899 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Sherwood, A. V., Grundy, F. J. & Henkin, T. M. T box riboswitches in actinobacteria: translational regulation via novel tRNA interactions. Proc. Natl Acad. Sci. USA 112, 1113–1118 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Chetnani, B. & Mondragon, A. Molecular envelope and atomic model of an anti-terminated glyQS T-box regulator in complex with tRNAGly. Nucleic Acids Res. 45, 8079–8090 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Fang, X., Michnicka, M., Zhang, Y., Wang, Y. X. & Nikonowicz, E. P. Capture and release of tRNA by the T-Loop receptor in the function of the T-Box riboswitch. Biochemistry 56, 3549–3558 (2017).

    CAS  PubMed  Google Scholar 

  30. Grundy, F. J. & Henkin, T. M. Kinetic analysis of tRNA-directed transcription antitermination of the Bacillus subtilis glyQS gene in vitro. J. Bacteriol. 186, 5392–5399 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, J. et al. Specific structural elements of the T-box riboswitch drive the two-step binding of the tRNA ligand. eLife 7, e39518 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Stagno, J. R. et al. Structures of riboswitch RNA reaction states by mix-and-inject XFEL serial crystallography. Nature 541, 242–246 (2017).

    CAS  PubMed  Google Scholar 

  33. Nozinovic, S. et al. The importance of helix P1 stability for structural pre-organization and ligand binding affinity of the adenine riboswitch aptamer domain. RNA Biol. 11, 655–656 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Peselis, A. & Serganov, A. ykkC riboswitches employ an add-on helix to adjust specificity for polyanionic ligands. Nat. Chem. Biol. 14, 887–894 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ren, A. et al. Structural and dynamic basis for low-affinity, high-selectivity binding of L-glutamine by the glutamine riboswitch. Cell Reports 13, 1800–1813 (2015).

    CAS  PubMed  Google Scholar 

  36. Doherty, E. A., Batey, R. T., Masquida, B. & Doudna, J. A. A universal mode of helix packing in RNA. Nat. Struct. Biol. 8, 339–343 (2001).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Sokoloski, J. E., Godfrey, S. A., Dombrowski, S. E. & Bevilacqua, P. C. Prevalence of syn nucleobases in the active sites of functional RNAs. RNA 17, 1775–1787 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Hemmes, P. R., Oppenheimer, L. & Jordan, F. Ultrasonic relaxation evaluation of the thermodynamics of syn-anti glycosidic isomerization in adenosine. J. Am. Chem. Soc. 96, 6023–6026 (1974).

    CAS  PubMed  Google Scholar 

  41. Dar, D. et al. Term-seq reveals abundant ribo-regulation of antibiotics resistance in bacteria. Science 352, aad9822 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Stamatopoulou, V. et al. Direct modulation of T-box riboswitch-controlled transcription by protein synthesis inhibitors. Nucleic Acids Res. 45, 10242–10258 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, J. et al. Identification of spermidine binding site in T-box riboswitch antiterminator RNA. Chem. Biol. Drug Des. 87, 182–189 (2016).

    CAS  PubMed  Google Scholar 

  44. Pleiss, J. A. & Uhlenbeck, O. C. Identification of thermodynamically relevant interactions between EF-Tu and backbone elements of tRNA. J. Mol. Biol. 308, 895–905 (2001).

    CAS  PubMed  Google Scholar 

  45. Taiji, M., Yokoyama, S. & Miyazawa, T. Transacylation rates of (aminoacyl)adenosine moiety at the 3’-terminus of aminoacyl transfer ribonucleic acid. Biochemistry 22, 3220–3225 (1983).

    CAS  PubMed  Google Scholar 

  46. Varani, G. & McClain, W. H. The G x U wobble base pair. A fundamental building block of RNA structure crucial to RNA function in diverse biological systems. EMBO Rep. 1, 18–23 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Masquida, B. & Westhof, E. On the wobble GoU and related pairs. RNA 6, 9–15 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Grundy, F. J., Moir, T. R., Haldeman, M. T. & Henkin, T. M. Sequence requirements for terminators and antiterminators in the T box transcription antitermination system: disparity between conservation and functional requirements. Nucleic Acids Res. 30, 1646–1655 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Gerdeman, M. S., Henkin, T. M. & Hines, J. V. Solution structure of the Bacillus subtilis T-box antiterminator RNA: seven nucleotide bulge characterized by stacking and flexibility. J. Mol. Biol. 326, 189–201 (2003).

    CAS  PubMed  Google Scholar 

  50. Grigg, J. C. et al. T box RNA decodes both the information content and geometry of tRNA to affect gene expression. Proc. Natl Acad. Sci. USA 110, 7240–7245 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Suddala, K. C. et al. Hierarchical mechanism of amino acid sensing by the T-box riboswitch. Nat. Commun. 9, 1896 (2018).

    PubMed  PubMed Central  Google Scholar 

  52. Zhang, K. et al. Structure of the 30 kDa HIV-1 RNA dimerization signal by a hybrid Cryo-EM, NMR, and molecular dynamics approach. Structure 26, 490–498.e3 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhang, J. & Ferre-D’Amare, A. R. Structure and mechanism of the T-box riboswitches. Wiley Interdiscip. Rev. RNA 6, 419–433 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Winkler, W. C., Grundy, F. J., Murphy, B. A. & Henkin, T. M. The GA motif: an RNA element common to bacterial antitermination systems, rRNA, and eukaryotic RNAs. RNA 7, 1165–1172 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Klein, D. J., Schmeing, T. M., Moore, P. B. & Steitz, T. A. The kink-turn: a new RNA secondary structure motif. EMBO J. 20, 4214–4221 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Van Treeck, B. & Parker, R. Emerging roles for intermolecular RNA-RNA interactions in RNP assemblies. Cell 174, 791–802 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Langdon, E. M. et al. mRNA structure determines specificity of a polyQ-driven phase separation. Science 360, 922–927 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Jain, A. & Vale, R. D. RNA phase transitions in repeat expansion disorders. Nature 546, 243–247 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Dethoff, E. A. et al. Pervasive tertiary structure in the dengue virus RNA genome. Proc. Natl Acad. Sci. USA 115, 11513–11518 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Leontis, N. B. & Westhof, E. Geometric nomenclature and classification of RNA base pairs. RNA 7, 499–512 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Franke, D. et al. ATSAS 2.8: a comprehensive data analysis suite for small-angle scattering from macromolecular solutions. J. Appl. Crystallogr. 50, 1212–1225 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Kalvari, I. et al. Rfam 13.0: shifting to a genome-centric resource for non-coding RNA families. Nucleic Acids Res. 46, D335–D342 (2018).

    CAS  PubMed  Google Scholar 

  63. Lorenz, R. et al. ViennaRNA Package 2.0. Algorithms Mol. Biol. 6, 26 (2011).

    PubMed  PubMed Central  Google Scholar 

  64. Crooks, G. E., Hon, G., Chandonia, J. M. & Brenner, S. E. WebLogo: a sequence logo generator. Genome Res. 14, 1188–1190 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Sampson, J. R. & Uhlenbeck, O. C. Biochemical and physical characterization of an unmodified yeast phenylalanine transfer RNA transcribed in vitro. Proc. Natl Acad. Sci. USA 85, 1033–1037 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhang, J. & Ferre-D’Amare, A. R. A flexible, scalable method for preparation of homogeneous aminoacylated tRNAs. Methods Enzymol. 549, 105–113 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Ferré-D’Amaré, A. R. & Doudna, J. A. Use of cis- and trans-ribozymes to remove 5′ and 3′ heterogeneities from milligrams of in vitro–transcribed RNA. Nucleic Acids Res. 24, 977–978 (1996).

    PubMed  PubMed Central  Google Scholar 

  68. Xiao, H., Murakami, H., Suga, H. & Ferré-D’Amaré, A. R. Structural basis of specific tRNA aminoacylation by a small in vitro selected ribozyme. Nature 454, 358–361 (2008).

    CAS  PubMed  Google Scholar 

  69. Hood, I. V. et al. Crystal structure of an adenovirus virus-associated RNA. Nat. Commun. 10, 2871 (2019).

    PubMed  PubMed Central  Google Scholar 

  70. Keller, S. et al. High-precision isothermal titration calorimetry with automated peak-shape analysis. Anal. Chem. 84, 5066–5073 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Zhao, H., Piszczek, G. & Schuck, P. SEDPHAT–a platform for global ITC analysis and global multi-method analysis of molecular interactions. Methods 76, 137–148 (2015).

    CAS  PubMed  Google Scholar 

  72. Zeng, C., Zhou, S., Bergmeier, S. C. & Hines, J. V. Factors that influence T box riboswitch efficacy and tRNA affinity. Bioorg. Med. Chem. 23, 5702–5708 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Yousef, M. R., Grundy, F. J. & Henkin, T. M. tRNA requirements for glyQS antitermination: a new twist on tRNA. RNA 9, 1148–1156 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Foadi, J. et al. Clustering procedures for the optimal selection of data sets from multiple crystals in macromolecular crystallography. Acta Crystallogr. D 69, 1617–1632 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Adams, P. et al. PHENIX: a comprehensive python-based system for macromolecular structure solution. Acta Cryst. D 66, 213–221 (2010).

    CAS  Google Scholar 

  76. McCoy, A. et al. Phaser crystallographic software. J. Appl. Cryst. 40, 658–674 (2007).

    CAS  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Chou, F. C., Sripakdeevong, P., Dibrov, S. M., Hermann, T. & Das, R. Correcting pervasive errors in RNA crystallography through enumerative structure prediction. Nat. Methods 10, 74–76 (2013).

    CAS  PubMed  Google Scholar 

  79. Chen, M. et al. Convolutional neural networks for automated annotation of cellular cryo-electron tomograms. Nat. Methods 14, 983–985 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Pettersen, E. F. et al. UCSF Chimera–a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    CAS  PubMed  Google Scholar 

  82. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D 74, 531–544 (2018).

    CAS  Google Scholar 

  83. Wang, J. et al. A method for helical RNA global structure determination in solution using small-angle x-ray scattering and NMR measurements. J. Mol. Biol. 393, 717–734 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Fang, X. et al. An unusual topological structure of the HIV-1 Rev response element. Cell 155, 594–605 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank I. Botos for computational support, J.R. Hogg, S. Ranganathan, G. Piszczek, D. Wu, J.C. Lee and M. Watson for support in fluorescence analyses; Y. He and N. Tjandra for fermentation support; R. Levine and D.-Y. Lee for MS support; W. Zhang and J. W. Szostak for a gift of Ir(III) Hexammine; M. Apostolidi for β-galactosidase assay protocols; CAS-Shanghai Science Research Center High-End User Project for preliminary cryo-EM data collection; and S.K. Buchanan, A. Ferré-D’Amaré, N. Baird, M. Lau, K. Suddala, C. Bou Nader, and J. M. Gordon for insightful discussions. This work was supported by the intramural research programs of NIDDK and NCI, an NIH DDIR Innovation Award to J.Z. and Y-X.W., NIH U54GM103297 and U54AI150470 (Center for HIV RNA Studies, CRNA), P41GM103832, R01GM079429 and S10OD021600 to W.C., and Fondatión Santé E515 and project “INSPIRED” (MIS/5002550) implemented under the Action “Reinforcement of the Research and Innovation Infrastructure”, funded by the Operational Programme “Competitiveness, Entrepreneurship and Innovation” (NSRF 2014-2020) and co-financed by Greece and the European Union (European Regional Development Fund), and a Fulbright Scholarship to C.S. V. S. is a supported by an IKY-Siemens 2017 Excellence Postdoctoral Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

S.L. and J.Z. designed experiments. S.L. prepared RNA samples, cocrystals, and samples for cryo-EM and SAXS, with the help of F.E.H, and performed in vitro assays. S.L. and J.Z. collected X-ray diffraction data, solved and refined the crystal structure, and analyzed SAXS data. Z.S. collected cryo-EM data, Z.S., G.D.P., K.Z., M.C., S.J.L. and W.C. performed cryo-EM data processing and modeling. J.L. performed phylogenetic analyses. V.S., N.G. and C.S. carried out in vivo experiments. L.F. and Y.-X.W. collected and processed SAXS data. All authors contributed to the preparation of the manuscript.

Corresponding authors

Correspondence to Wah Chiu or Jinwei Zhang.

Ethics declarations

Competing interests

Authors declare no competing interests.

Additional information

Peer review information Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Secondary structures and conservation analyses of glycyl T-box riboswitches.

a,b, Secondary structures of G. kaustophilus glyQ and B. subtilis glyQS T-box riboswitch. Glycine-specific T-boxes lack the stem II and stem IIA/B pseudoknot structures. Conserved nucleotides are highlighted, based on previous reports supplemented by new phylogenetic analysis (Fig. 1; Methods). Previous sequence annotations of the G. kaustophilus glyQ T-box had omitted a 5′ ssRNA leader that precedes stem I in all validated T-boxes, which is now restored. The probable transcription start site, 17 nts upstream of stem I, was identified using prokaryotic promoter prediction algorithms. Nucleotide numbering is thus offset by +17 relative to previous reports. c, Sequence conservation of the T-box discriminator region based on G. kaustophilus glyQ T-box. The split patterns show that the intercalating G130 is at the center of a 5′-AR(U/A)-3′ motif (middle). This motif is shifted 1 nt to the left when there is no G in position 130 (bottom). In this case, a moderately conserved G is predominant in position 129, and two pyrimidines are present in positions 130 and 131. Assuming that G129 is the intercalating nucleotide equivalent to G130 in the middle panel, one of these pyrimidines (nt 130 or 131) may adopt an extrahelical conformation to account for the motif shift.

Extended Data Fig. 2 Mutational analysis of T-box discriminator-tRNA interactions.

a, Secondary structures of wild-type, mutant, and truncated T-box discriminators. Deletions are indicated by red boxes. b, Electrophoretic mobility shift assay (EMSA) analysis of the constructs shown in a, showing the requirement of stem III and flanking purines for tRNA binding. The antiterminator (discriminator without stem III and its flanking purines; Δ3 mutant) is prone to dimerization. c, tRNA variants used that carry various 3′ chemical modifications. Only the terminal tA76 is shown. d, EMSA analysis of constructs in c, showing that binding is selective for uncharged tRNA. e, Quantitation of d and comparison with previously reported in vitro transcription readthrough data of the same tRNA variants. The values and error bars represent mean and s.d., n = 3 biologically independent samples.

Extended Data Fig. 3 Representative X-ray crystallographic electron density maps.

a, Composite simulated anneal-omit 2FoFc electron density calculated using the final model (1.0 s.d.) superimposed with the final refined model. bd, Portions of the map showing tRNA-T-box discriminator coaxial stacking (b), encapsulation of tRNA 3′-end by the discriminator (c), and long-range interactions between stem III 5′ flanking purines and the T-box bulge (d). Note the density fusion as a result of nucleobase-ribose packing interactions between A129 and G161.

Extended Data Fig. 4 In vitro transcription termination-readthrough assay and in vivo β-gal assay.

a, Representative raw data of in vitro transcription termination-readthrough assay using wild-type B. subtilis T-box riboswitch. The rates of fluorescence increase between 34 and 180 min (segments with trendlines) report the production of readthrough transcripts. b, Quantitation of data in a. Rates of fluorescence increase (slopes) were subsequently normalized to that of the reference in the presence of NTP but absence of tRNA (green data points) and reported in Figs. 2h, 3e and 4f. c, Validation of fluorescence-based readthrough assay in a and b with subsequent, conventional gel-based analysis of the same samples. Addition of the uncharged tRNA led to significantly increased transcription readthrough. d, Scheme of in vivo gene expression assay using the G. kaustophilus glyQ T-box riboswitch transcriptionally fused with lacZ. e, Relative β-gal activity of wild-type and mutant T-boxes under glycine-replete and glycine-starvation conditions, normalized to wild-type T-box-containing strain grown in minimal media supplemented with glycine. The values and error bars represent mean and s.d., n = 3 biologically independent samples.

Extended Data Fig. 5 Comparison of the T-box tandem A-minor latch with the A1492-A1493-G530 latch in the ribosome A site.

a, The A128-A129 latch reinforces the functionally important tRNA (green)-helix A1 (blue) stacking interface. A128 and A129 form a continuous adenosine stack. b and c, The stacked A128 and A129 engage extensive hydrogen bonds with the minor groove, reinforce tRNA-T-box base-pairing and stacking, and “staple” the two RNAs together. d, In the ribosome A site, A1492 and A1493 similarly reinforce the intermolecular codon-anticodon duplex via tandem, stacked A-minor interactions in conjunction with G530. e and f, Hydrogen-bond patterns in the ribosome A site resemble those in the T-box (b and c).

Extended Data Fig. 6 Intermolecular interface of the T-box discriminator-tRNA complex.

a, Solvent-accessible surface colored according to area buried from light blue or white (no burial) to red (>25 Å2 per atom). b, Open-book view of the binding interface. The lower inset shows the extensive burial of tRNA tA76, particularly its Watson−Crick edge (N6-N1-C2) and both 2′-OH and 3′-OH. c,d, Solvent-accessible surface area buried per residue for tRNA (c) and discriminator (d).

Extended Data Fig. 7 Effect of the G•U wobble pair on tRNA aminoacylation sensing and comparison of steric sieves in the T-box and ribosome-RelA complex.

a, A modeled tRNA 3′-glycyl moiety strongly clashes with the U185 nucleobase of the G•U wobble pair. b, Modeled Watson–Crick pair (C185, white) still clashes with the tRNA 3′-glycyl moiety, albeit to a lesser extent than the G•U wobble pair (a). c,d, Comparison of the steric sieves in the T-box (c) snd RelA–ribosome complex (d). Solid green lines indicate inter-atomic distances in Å. The RelA–ribosome complex structure is based on PDB 5IQR.

Extended Data Fig. 8 A conserved G•U wobble pair enhances stacking with its neighboring base pair both in the T-box discriminator and in the tRNA T-loop.

a, Through local helix underwinding, helix A2 terminal G•U wobble pair produces exceptionally large nucleobase overlap areas and enhances stacking with the penultimate C-G pair. b, Reduced nucleobase overlap areas between a modeled G-C pair and the penultimate C-G pair. c, The G•U wobble pair is reminiscent of the conserved G49•U65 wobble pair found in the tRNAGly T-loop in the same complex. d, For comparison, the penultimate C-G pair stacks with its neighboring G-C pair with less than half of the total overlap area (5.5 Å2 versus 13.9 Å2). Overlap areas (in Å2) between stacked nucleobases were calculated with 3DNA.

Extended Data Fig. 9 Cryo-EM single particle analysis (SPA) workflow of full-length B. subtilis T-box−tRNA complex.

a, 3D classification yielded two major classes (black boxes) that were combined for auto refinement. b, Final reconstruction. c, FSC curve showing 4.9-Å resolution at 0.143 cut-off. d, Euler angle distribution of the final reconstruction.

Extended Data Fig. 10 Relion 3D classification and local resolution of the full-length B. subtilis T-box−tRNA complex.

3D classification of the complex converged to three maps. Superposition of the tRNA density in three maps revealed motions of the T-box relative to tRNA as indicated by the arrows. This flexibility of the T-box RNA is the major limitation that prevented cryo-EM reconstruction from achieving higher resolutions. Resmap analysis shows that the tRNA was better resolved at ~4-Å resolution (upper right).

Supplementary information

Reporting Summary

Supplementary Video 1

360° view of the 2.66-Å resolution cocrystal structure of the T-box discriminator in complex with an uncharged tRNA.

Supplementary Video 2

Animation of the 4.9-Å resolution Cryo-EM structure of the full-length T-box riboswitch-tRNA complex. Multiple zoom-in views display the match between the model and the densities including the three separate tRNA-mRNA interfaces and the better resolved regions of the map in the tRNA.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, S., Su, Z., Lehmann, J. et al. Structural basis of amino acid surveillance by higher-order tRNA-mRNA interactions. Nat Struct Mol Biol 26, 1094–1105 (2019). https://doi.org/10.1038/s41594-019-0326-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0326-7

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