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 for tRNA modification by Elp3 from Dehalococcoides mccartyi

Abstract

During translation elongation, decoding is based on the recognition of codons by corresponding tRNA anticodon triplets. Molecular mechanisms that regulate global protein synthesis via specific base modifications in tRNA anticodons are receiving increasing attention. The conserved eukaryotic Elongator complex specifically modifies uridines located in the wobble base position of tRNAs. Mutations in Elongator subunits are associated with certain neurodegenerative diseases and cancer. Here we present the crystal structure of D. mccartyi Elp3 (DmcElp3) at 2.15-Å resolution. Our results reveal an unexpected arrangement of Elp3 lysine acetyltransferase (KAT) and radical S-adenosyl methionine (SAM) domains, which share a large interface and form a composite active site and tRNA-binding pocket, with an iron–sulfur cluster located in the dimerization interface of two DmcElp3 molecules. Structure-guided mutagenesis studies of yeast Elp3 confirmed the relevance of our findings for eukaryotic Elp3s and should aid in understanding the cellular functions and pathophysiological roles of Elongator.

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

Figure 1: Crystal structure of DmcElp3.
Figure 2: Dimerization of DmcElp3.
Figure 3: DmcElp3 tRNA interaction.
Figure 4: RNase protection assays and tRNA–DmcElp3 model.
Figure 5: Model of the individual steps of the Elp3 modification reaction.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Fredrick, K. & Ibba, M. How the sequence of a gene can tune its translation. Cell 141, 227–229 (2010).

    Article  CAS  Google Scholar 

  2. Pechmann, S. & Frydman, J. Evolutionary conservation of codon optimality reveals hidden signatures of cotranslational folding. Nat. Struct. Mol. Biol. 20, 237–243 (2013).

    Article  CAS  Google Scholar 

  3. Komar, A.A. A pause for thought along the co-translational folding pathway. Trends Biochem. Sci. 34, 16–24 (2009).

    Article  CAS  Google Scholar 

  4. Thanaraj, T.A. & Argos, P. Ribosome-mediated translational pause and protein domain organization. Protein Sci. 5, 1594–1612 (1996).

    Article  CAS  Google Scholar 

  5. Rezgui, V.A. et al. tRNA tKUUU, tQUUG, and tEUUC wobble position modifications fine-tune protein translation by promoting ribosome A-site binding. Proc. Natl. Acad. Sci. USA 110, 12289–12294 (2013).

    Article  CAS  Google Scholar 

  6. 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  Google Scholar 

  7. Nedialkova, D.D. & Leidel, S.A. Optimization of codon translation rates via tRNA modifications maintains proteome integrity. Cell 161, 1606–1618 (2015).

    Article  CAS  Google Scholar 

  8. Glatt, S. & Müller, C.W. Structural insights into Elongator function. Curr. Opin. Struct. Biol. 23, 235–242 (2013).

    Article  CAS  Google Scholar 

  9. Huang, B., Johansson, M.J.O. & Byström, A.S. An early step in wobble uridine tRNA modification requires the Elongator complex. RNA 11, 424–436 (2005).

    Article  CAS  Google Scholar 

  10. Selvadurai, K., Wang, P., Seimetz, J. & Huang, R.H. Archaeal Elp3 catalyzes tRNA wobble uridine modification at C5 via a radical mechanism. Nat. Chem. Biol. 10, 810–812 (2014).

    Article  CAS  Google Scholar 

  11. Chen, C., Huang, B., Anderson, J.T. & Byström, A.S. Unexpected accumulation of ncm5U and ncm5s2U in a trm9 mutant suggests an additional step in the synthesis of mcm5U and mcm5s2U. PLoS One 6, e20783 (2011).

    Article  CAS  Google Scholar 

  12. Esberg, A., Huang, B., Johansson, M.J.O. & Byström, A.S. Elevated levels of two tRNA species bypass the requirement for elongator complex in transcription and exocytosis. Mol. Cell 24, 139–148 (2006).

    Article  CAS  Google Scholar 

  13. Beck, M. et al. The quantitative proteome of a human cell line. Mol. Syst. Biol. 7, 549 (2011).

    Article  Google Scholar 

  14. Walker, J. et al. Role of elongator subunit Elp3 in Drosophila melanogaster larval development and immunity. Genetics 187, 1067–1075 (2011).

    Article  CAS  Google Scholar 

  15. Chen, C., Tuck, S. & Byström, A.S. Defects in tRNA modification associated with neurological and developmental dysfunctions in Caenorhabditis elegans elongator mutants. PLoS Genet. 5, e1000561 (2009).

    Article  Google Scholar 

  16. Simpson, C.L. et al. Variants of the elongator protein 3 (ELP3) gene are associated with motor neuron degeneration. Hum. Mol. Genet. 18, 472–481 (2009).

    Article  CAS  Google Scholar 

  17. Mehlgarten, C. et al. Elongator function in tRNA wobble uridine modification is conserved between yeast and plants. Mol. Microbiol. 76, 1082–1094 (2010).

    Article  CAS  Google Scholar 

  18. Chen, Y.T. et al. Loss of mouse Ikbkap, a subunit of elongator, leads to transcriptional deficits and embryonic lethality that can be rescued by human IKBKAP. Mol. Cell. Biol. 29, 736–744 (2009).

    Article  CAS  Google Scholar 

  19. Slaugenhaupt, S.A. et al. Tissue-specific expression of a splicing mutation in the IKBKAP gene causes familial dysautonomia. Am. J. Hum. Genet. 68, 598–605 (2001).

    Article  CAS  Google Scholar 

  20. Wittschieben, B.O. et al. A novel histone acetyltransferase is an integral subunit of elongating RNA polymerase II holoenzyme. Mol. Cell 4, 123–128 (1999).

    Article  CAS  Google Scholar 

  21. Paraskevopoulou, C., Fairhurst, S.A., Lowe, D.J., Brick, P. & Onesti, S. The Elongator subunit Elp3 contains a Fe4S4 cluster and binds S-adenosylmethionine. Mol. Microbiol. 59, 795–806 (2006).

    Article  CAS  Google Scholar 

  22. Lemmens, R., Moore, M.J., Al-Chalabi, A., Brown, R.H. Jr. & Robberecht, W. RNA metabolism and the pathogenesis of motor neuron diseases. Trends Neurosci. 33, 249–258 (2010).

    Article  CAS  Google Scholar 

  23. Xu, H. et al. Dimerization of elongator protein 1 is essential for Elongator complex assembly. Proc. Natl. Acad. Sci. USA 112, 10697–10702 (2015).

    Article  CAS  Google Scholar 

  24. Dong, C. et al. The Elp2 subunit is essential for elongator complex assembly and functional regulation. Structure 23, 1078–1086 (2015).

    Article  CAS  Google Scholar 

  25. Glatt, S. et al. The Elongator subcomplex Elp456 is a hexameric RecA-like ATPase. Nat. Struct. Mol. Biol. 19, 314–320 (2012).

    Article  CAS  Google Scholar 

  26. Lin, Z. et al. Crystal structure of elongator subcomplex Elp4-6. J. Biol. Chem. 287, 21501–21508 (2012).

    Article  CAS  Google Scholar 

  27. Glatt, S. et al. Structure of the Kti11/Kti13 heterodimer and its double role in modifications of tRNA and eukaryotic elongation factor 2. Structure 23, 149–160 (2015).

    Article  CAS  Google Scholar 

  28. Kolaj-Robin, O., McEwen, A.G., Cavarelli, J. & Séraphin, B. Structure of the Elongator cofactor complex Kti11/Kti13 provides insight into the role of Kti13 in Elongator-dependent tRNA modification. FEBS J. 282, 819–833 (2015).

    Article  CAS  Google Scholar 

  29. Svejstrup, J.Q. Elongator complex: how many roles does it play? Curr. Opin. Cell Biol. 19, 331–336 (2007).

    Article  CAS  Google Scholar 

  30. Versées, W., De Groeve, S. & Van Lijsebettens, M. Elongator, a conserved multitasking complex? Mol. Microbiol. 76, 1065–1069 (2010).

    Article  Google Scholar 

  31. Glatt, S., Séraphin, B. & Müller, C.W. Elongator: transcriptional or translational regulator? Transcription 3, 273–276 (2012).

    Article  Google Scholar 

  32. Karlsborn, T., Tükenmez, H., Chen, C. & Byström, A.S. Familial dysautonomia (FD) patients have reduced levels of the modified wobble nucleoside mcm5s2U in tRNA. Biochem. Biophys. Res. Commun. 454, 441–445 (2014).

    Article  CAS  Google Scholar 

  33. Najmabadi, H. et al. Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature 478, 57–63 (2011).

    Article  CAS  Google Scholar 

  34. Löffler, F.E. et al. Dehalococcoides mccartyi gen. nov., sp. nov., obligately organohalide-respiring anaerobic bacteria relevant to halogen cycling and bioremediation, belong to a novel bacterial class, Dehalococcoidia classis nov., order Dehalococcoidales ord. nov. and family Dehalococcoidaceae fam. nov., within the phylum Chloroflexi. Int. J. Syst. Evol. Microbiol. 63, 625–635 (2013).

    Article  Google Scholar 

  35. Seshadri, R. et al. Genome sequence of the PCE-dechlorinating bacterium Dehalococcoides ethenogenes. Science 307, 105–108 (2005).

    Article  CAS  Google Scholar 

  36. Boal, A.K. et al. Structural basis for methyl transfer by a radical SAM enzyme. Science 332, 1089–1092 (2011).

    Article  CAS  Google Scholar 

  37. Benítez-Páez, A., Villarroya, M. & Armengod, M.E. The Escherichia coli RlmN methyltransferase is a dual-specificity enzyme that modifies both rRNA and tRNA and controls translational accuracy. RNA 18, 1783–1795 (2012).

    Article  Google Scholar 

  38. Schwalm, E.L., Grove, T.L., Booker, S.J. & Boal, A.K. Crystallographic capture of a radical S-adenosylmethionine enzyme in the act of modifying tRNA. Science 352, 309–312 (2016).

    Article  CAS  Google Scholar 

  39. Dyda, F., Klein, D.C. & Hickman, A.B. GCN5-related N-acetyltransferases: a structural overview. Annu. Rev. Biophys. Biomol. Struct. 29, 81–103 (2000).

    Article  CAS  Google Scholar 

  40. Creppe, C. et al. Elongator controls the migration and differentiation of cortical neurons through acetylation of alpha-tubulin. Cell 136, 551–564 (2009).

    Article  CAS  Google Scholar 

  41. Mis´kiewicz, K. et al. ELP3 controls active zone morphology by acetylating the ELKS family member Bruchpilot. Neuron 72, 776–788 (2011).

    Article  Google Scholar 

  42. Sweeney, W.V. & Rabinowitz, J.C. Proteins containing 4Fe-4S clusters: an overview. Annu. Rev. Biochem. 49, 139–161 (1980).

    Article  CAS  Google Scholar 

  43. Lanciano, P. et al. New method for the spin quantitation of [4Fe-4S]+ clusters with S = 3/2: application to the FS0 center of the NarGHI nitrate reductase from Escherichia coli. J. Phys. Chem. B 111, 13632–13637 (2007).

    Article  CAS  Google Scholar 

  44. Conover, R.C. et al. Spectroscopic characterization of the novel iron-sulfur cluster in Pyrococcus furiosus ferredoxin. J. Biol. Chem. 265, 8533–8541 (1990).

    CAS  PubMed  Google Scholar 

  45. Bruska, M.K., Stiebritz, M.T. & Reiher, M. Analysis of differences in oxygen sensitivity of Fe-S clusters. Dalton Trans. 42, 8729–8735 (2013).

    Article  CAS  Google Scholar 

  46. Dong, M. et al. Dph3 is an electron donor for Dph1-Dph2 in the first step of eukaryotic diphthamide biosynthesis. J. Am. Chem. Soc. 136, 1754–1757 (2014).

    Article  CAS  Google Scholar 

  47. Jablonowski, D., Zink, S., Mehlgarten, C., Daum, G. & Schaffrath, R. tRNAGlu wobble uridine methylation by Trm9 identifies Elongator's key role for zymocin-induced cell death in yeast. Mol. Microbiol. 59, 677–688 (2006).

    Article  CAS  Google Scholar 

  48. Nelissen, H. et al. The elongata mutants identify a functional Elongator complex in plants with a role in cell proliferation during organ growth. Proc. Natl. Acad. Sci. USA 102, 7754–7759 (2005).

    Article  CAS  Google Scholar 

  49. Greenwood, C., Selth, L.A., Dirac-Svejstrup, A.B. & Svejstrup, J.Q. An iron-sulfur cluster domain in Elp3 important for the structural integrity of elongator. J. Biol. Chem. 284, 141–149 (2009).

    Article  CAS  Google Scholar 

  50. Okada, Y., Yamagata, K., Hong, K., Wakayama, T. & Zhang, Y. A role for the elongator complex in zygotic paternal genome demethylation. Nature 463, 554–558 (2010).

    Article  CAS  Google Scholar 

  51. Wittschieben, B.O., Fellows, J., Du, W., Stillman, D.J. & Svejstrup, J.Q. Overlapping roles for the histone acetyltransferase activities of SAGA and elongator in vivo. EMBO J. 19, 3060–3068 (2000).

    Article  CAS  Google Scholar 

  52. Wang, J. et al. Recent advances in radical SAM enzymology: new structures and mechanisms. ACS Chem. Biol. 9, 1929–1938 (2014).

    Article  CAS  Google Scholar 

  53. Clements, A. et al. Structural basis for histone and phosphohistone binding by the GCN5 histone acetyltransferase. Mol. Cell 12, 461–473 (2003).

    Article  CAS  Google Scholar 

  54. Rojas, J.R. et al. Structure of Tetrahymena GCN5 bound to coenzyme A and a histone H3 peptide. Nature 401, 93–98 (1999).

    Article  CAS  Google Scholar 

  55. Poux, A.N. & Marmorstein, R. Molecular basis for Gcn5/PCAF histone acetyltransferase selectivity for histone and nonhistone substrates. Biochemistry 42, 14366–14374 (2003).

    Article  CAS  Google Scholar 

  56. Sekine, S., Nureki, O., Shimada, A., Vassylyev, D.G. & Yokoyama, S. Structural basis for anticodon recognition by discriminating glutamyl-tRNA synthetase. Nat. Struct. Biol. 8, 203–206 (2001).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  58. de la Fortelle, E. & Bricogne, G. Maximum-likelihood heavy-atom parameter refinement for multiple isomorphous replacement and multiwavelength anomalous diffraction methods. Macromolecular Crystallography Part A 276, 472–494 (1997).

    Article  CAS  Google Scholar 

  59. Adams, P.D. et al. PHENIX: building new software for automated crystallographic structure determination. Acta Crystallogr. D Biol. Crystallogr. 58, 1948–1954 (2002).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  61. Davis, I.W. et al. MolProbity: all-atom contacts and structure validation for proteins and nucleic acids. Nucleic Acids Res. 35, W375–W383 (2007).

    Article  Google Scholar 

  62. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  Google Scholar 

  63. Yang, J. et al. The I-TASSER Suite: protein structure and function prediction. Nat. Methods 12, 7–8 (2015).

    Article  CAS  Google Scholar 

  64. Kelley, L.A. & Sternberg, M.J. Protein structure prediction on the Web: a case study using the Phyre server. Nat. Protoc. 4, 363–371 (2009).

    Article  CAS  Google Scholar 

  65. Silakov, A. et al. Characterization of a cross-linked protein-nucleic acid substrate radical in the reaction catalyzed by RlmN. J. Am. Chem. Soc. 136, 8221–8228 (2014).

    Article  CAS  Google Scholar 

  66. van Zundert, G.C. et al. The HADDOCK2.2 web server: user-friendly integrative modeling of biomolecular complexes. J. Mol. Biol. 428, 720–725 (2016).

    Article  CAS  Google Scholar 

  67. Knop, M. et al. Epitope tagging of yeast genes using a PCR-based strategy: more tags and improved practical routines. Yeast 15 10B, 963–972 (1999).

    Article  CAS  Google Scholar 

  68. Mumberg, D., Müller, R. & Funk, M. Yeast vectors for the controlled expression of heterologous proteins in different genetic backgrounds. Gene 156, 119–122 (1995).

    Article  CAS  Google Scholar 

  69. Frohloff, F., Fichtner, L., Jablonowski, D., Breunig, K.D. & Schaffrath, R. Saccharomyces cerevisiae Elongator mutations confer resistance to the Kluyveromyces lactis zymocin. EMBO J. 20, 1993–2003 (2001).

    Article  CAS  Google Scholar 

  70. Boivin, S., Kozak, S. & Meijers, R. Optimization of protein purification and characterization using Thermofluor screens. Protein Expr. Purif. 91, 192–206 (2013).

    Article  CAS  Google Scholar 

  71. Franke, D., Jeffries, C.M. & Svergun, D.I. Correlation Map, a goodness-of-fit test for one-dimensional X-ray scattering spectra. Nat. Methods 12, 419–422 (2015).

    Article  CAS  Google Scholar 

  72. Petoukhov, M.V. et al. New developments in the ATSAS program package for small-angle scattering data analysis. J. Appl. Crystallogr. 45, 342–350 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Lin (Cornell University) for providing the Azotobacter vinelandii isc operon–containing plasmid, G. Sawers (Martin-Luther-University Halle) for providing genomic DNA from D. mccartyi, G. Fritz (University of Freiburg) for discussion and H. Grötsch for support. We acknowledge support from the EMBL Heidelberg Crystallization Platform. We also acknowledge access and support provided by the EMBL/ESRF Joint Structural Biology Group at ESRF beamlines. R.Z. and K.D.B. acknowledge funding from the German Science Foundation (SFB 648). EPR work was supported by TGE RPE FR3443 (F. Baymann). This work was also supported by the Ligue contre le Cancer (Equipe labellisée 2014) (B.S.), the Centre National pour la Recherche Scientifique (B.S.), the CERBM–IGBMC, the project Elongator from the Agence Nationale pour la Recherche (grant ANR-13-BSV8-0005-01) (B.S.) and grant ANR-10-LABX-0030-INRT managed under the program Investissements d'Avenir ANR-10-IDEX-0002-02 (B.S.).

Author information

Authors and Affiliations

Authors

Contributions

S.G. performed the biochemical, biophysical and crystallographic analyses of DmcElp3 with the help of T.-Y.L. R.Z. and O.F.O. characterized mutant phenotypes in yeast. O.K.-R. and V.T. carried out anaerobic purifications, acetylation assays and spectroscopic analyses. F. Baymann. performed EPR measurements. F. Baudin performed RNase protection assays. A.G. collected and analyzed SAXS data. S.G., O.K.-R., B.S., K.D.B. and C.W.M. designed experiments and analyzed the data. S.G. and C.W.M. wrote the manuscript with input from all other authors.

Corresponding author

Correspondence to Christoph W Müller.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Topology and structural features of DmcElp3.

(a) Anomalous difference Fourier map of DmcElp3 collected at the selenium absorption edge shown at 5 I/σ. Selenomethionine residues are shown in ball and sticks representation. (b) Topology of DmcElp3 indicating secondary structure elements. (c) Secondary structure elements are shown above the amino acid sequence showing only residues present in the structural model and highlighting cysteine residues involved in iron-sulfur cluster or zinc coordination. (d) Stereo figure showing the different indicated substrate peptides bound to Gcn5 resulting from a superimposition of Gcn5 with the KAT domain of DmcElp3. Used structures were 1Q2C (H4), 1QSN (H3), 1PUA (pH3) and 1Q2D (p53) (Clements, A. et al., Mol Cell 12, 461–473, 2003; Rojas, J.R. et al., Nature 401, 93-98; Poux, A.N. and Marmorstein, R., Biochemistry 42, 14366-14374).

Supplementary Figure 2 Analyses of purified DmcElp3.

(a) UV/Vis absorption spectra of anaerobically purified DmcElp3 showing the broad absorption band characteristic of [4Fe-4S]2+ cluster at 410 nm. (b) EPR spectra of anaerobically (upper traces, protein concentration 7.5 mg/ml) and aerobically (lower traces, protein concentration 10.6 mg/ml) purified DmcElp3 protein recorded at 6 K with a microwave power of 128 mW and a modulation amplitude of 0.00303 T (left panel) and at 50 K with a microwave power of 6.4 mW and a modulation amplitude of 0.001 T (right panel) on a Bruker Elexsys E500 X-band spectrometer fitted with an Oxford Instrument He-cryostat ESR900 and temperature control system. Microwave frequency was 9.48 GHz. (c) Gelfiltration profiles of indicated purified DmcElp3 mutants. (d) Thermofluor analyses of indicated monomeric and dimeric DmcElp3 mutants. (e) Fitting of experimental scattering data (dots) for Elp3 monomer (red, 0.58 mg/ml) and dimer (black, 0.54 mg/ml) to theoretical scattering curves calculated from the structure (lines) show that the model predicts the low-resolution features of both the monomer and the dimer in solution. Furthermore, the Guinier analysis in the inset shows a good agreement in terms of radius of gyration (Rg) between theoretical and experimental data. Fitting calculated using CRYSOL (ATSAS suite). Theoretical Rg Dimer =3.33 nm. Theoretical Rg Monomer=2.22 nm. (f) Titration of acetyl-CoA and SAM onto Elp3 monomer and dimer. No large conformational change is observed upon addition of acetyl-CoA or SAM, with the protein maintaining a conformation compatible with that of the crystal structure. The curves displayed are extrapolations to infinite dilution calculated using PRIMUS (ATSAS suite) from concentration series, in order to minimize inter-particle effects (see methods).

Supplementary Figure 3 tRNA binding of DmcElp3 and structural surface analyses.

(a) Unspecific acetylation of DmcElp3 and unrelated proteins (human BTG2, yeast Kti13 and E.coli NorW) in the presence of Na2O4S2 detected by using radioactive labelled (C14) acetyl-CoA. (b) EMSA analysis of monomeric and dimeric DmcElp3 using tRNAGlu from D. mccartyi (left) and S. cerevisiae (right). (c) Structure of DmcElp3 shown in cartoon representation (top), surface representation showing conservation scores of 394 unique sequences from ConSurf analyses (www.consurf.tau.ac.il/) (middle) and surface representation showing surface charge distribution from APBS analysis (bottom). The conserved basic tRNA binding cavity is indicated by a dotted circle.

Supplementary Figure 4 tRNA binding and phenotypes of additional DmcElp3 mutants.

(a,b) EMSA analysis of indicated point mutants using radioactively labelled tRNA and increasing amounts of protein (1.6, 7.5, 15 μM) on a 6% native gel. Samples were also analyzed using denaturing SDS-PAGE as loading control. (c,d) Phenotypes of yeast strains carrying the indicated Scelp3 mutant alleles using γ-toxin and tRNA suppression assays. γ-toxin and SUP4 suppression assays are described in the Online Methods.

Supplementary Figure 5 Comparing crystal structures of full-length and 390–407(GSGSG) DmcElp3.

(a) Comparison between binding affinities of full length DmcElp3 and 390-407GSGSG DmcElp3 using EMSA analysis of indicated point mutants using radioactively labelled tRNA and increasing amounts of protein (0.9, 1.8, 3.7, 7.5, 15 μM) on a 6% native gel. Samples were also analyzed using denaturing SDS-PAGE as loading control. (b) 2mFo-Fc density of full length DmcElp3 (left) and 390-407GSGSG DmcElp3 (right) shown at 1 I/σ. (c) Close up of the acetyl-CoA binding site showing 2mFo-Fc density for the acetyl-CoA blocking loop and the 5GS linker region (left, middle). Model of fitted CoA (green) bound to the acetyl-CoA binding site of 390-407GSGSG using CoA from 1QSR (right). 2mFo-Fc density with omitted acetyl-CoA is shown in green.

Supplementary Figure 6 Localization of known Elp3 mutations in the DmcElp3 structure.

(a) Summary of previously tested Elp3 mutations. Tested organisms (D. melanogaster (Dm), S. cerevisiae (Sc), A. thaliana (At), H. sapiens (Hs), M. infernus (Min), M. jannaschii (Mj), M. musculus (Mm)), original mutations and equivalent positions are listed. Identical residues are in bold and cysteine residues involved in zinc coordination are underlined. All mutants are mapped on the DmcElp3 structure and are highlighted (blue/green) using balls and sticks representation. (b) Summary of identified alterations of human Elp3 from ICGC (www.icgc.com) and TCGA (www.cancergenome.nih.org). Equivalent positions are highlighted red and using balls and sticks representation. (c) Complementation phenotypes from the indicated genomic integrated Scelp3 mutants using γ-toxin and tRNA suppression assays. Tested mutations related to neurodegenerative diseases in (a) are labelled with an asterisk (*) and mutations identified in cancer patients in (b) are labelled by two asterisks (**).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6, Supplementary Table 1 and Supplementary Notes 1 and 2 (PDF 2467 kb)

Supplementary Data Set 1

Uncropped gels and autoradiographs (PDF 2030 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Glatt, S., Zabel, R., Kolaj-Robin, O. et al. Structural basis for tRNA modification by Elp3 from Dehalococcoides mccartyi. Nat Struct Mol Biol 23, 794–802 (2016). https://doi.org/10.1038/nsmb.3265

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3265

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