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 Cfr-mediated antimicrobial resistance and mechanisms to evade it

Abstract

The bacterial ribosome is an essential drug target as many clinically important antibiotics bind and inhibit its functional centers. The catalytic peptidyl transferase center (PTC) is targeted by the broadest array of inhibitors belonging to several chemical classes. One of the most abundant and clinically prevalent resistance mechanisms to PTC-acting drugs in Gram-positive bacteria is C8-methylation of the universally conserved A2503 nucleobase by Cfr methylase in 23S ribosomal RNA. Despite its clinical importance, a sufficient understanding of the molecular mechanisms underlying Cfr-mediated resistance is currently lacking. Here, we report a set of high-resolution structures of the Cfr-modified 70S ribosome containing aminoacyl- and peptidyl-transfer RNAs. These structures reveal an allosteric rearrangement of nucleotide A2062 upon Cfr-mediated methylation of A2503 that likely contributes to the reduced potency of some PTC inhibitors. Additionally, we provide the structural bases behind two distinct mechanisms of engaging the Cfr-methylated ribosome by the antibiotics iboxamycin and tylosin.

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: T. thermophilus HB27 strain expressing Cfr-like methylase.
Fig. 2: Electron density maps of C2,C8-dimethylated (top) and C2-methylated (bottom) A2503 residue of the 23S rRNA in T. thermophilus 70S ribosome.
Fig. 3: Structure of the Cfr-modified 70S ribosome.
Fig. 4: Structural basis for Cfr-mediated resistance to PTC-acting antibiotics.
Fig. 5: Structure of IBX bound to the Cfr-methylated 70S ribosome.
Fig. 6: Comparison of the structures of TYL bound to the Cfr-modified and WT 70S ribosomes.

Similar content being viewed by others

Data availability

Coordinates and structure factors were deposited in the RCSB Protein Data Bank with accession codes: 8G29 for the A2503-C2,C8-dimethylated T. thermophilus 70S ribosome in complex with mRNA, aminoacylated A-site Phe-NH-tRNAPhe, aminoacylated P-site fMet-NH-tRNAiMet and deacylated E-site tRNAPhe; 8G2A for the A2503-C2,C8-dimethylated T. thermophilus 70S ribosome in complex with mRNA, aminoacylated A-site Phe-NH-tRNAPhe, peptidyl P-site fMTHSMRC-NH-tRNAiMet and deacylated E-site tRNAPhe. 8G2B for the A2503-C2,C8-dimethylated T. thermophilus 70S ribosome in complex with mRNA, deacylated A-site tRNAPhe, aminoacylated P-site fMet-NH-tRNAiMet, deacylated E-site tRNAPhe and iboxamycin; 8G2C for the A2503-C2,C8-dimethylated T. thermophilus 70S ribosome in complex with mRNA, aminoacylated A-site Phe-NH-tRNAPhe, aminoacylated P-site fMet-NH-tRNAiMet, deacylated E-site tRNAPhe and tylosin; 8G2D for the wild-type T. thermophilus 70S ribosome in complex with mRNA, deacylated A-site tRNAPhe, deacylated P-site tRNAiMet, deacylated E-site tRNAPhe and tylosin. All previously published structures that were used in this work for structural comparisons were retrieved from the RCSB Protein Data Bank: PDB entries 6XHW, 8CVL, 7LVK, 7RQE, 7S1G, 7S1I, 5DOY, 5VP2, 4V7V, 7RQ8, 1K9M, 1KD1, 1K8A. No sequence data were generated in this study. Analyzed protein sequences are presented with their corresponding accession numbers in the phylogenetic tree (Supplementary Fig. 1) for retrieval from the NCBI protein database. Source data are provided with this paper.

References

  1. Wilson, D. N. The A-Z of bacterial translation inhibitors. Crit. Rev. Biochem. Mol. Biol. 44, 393–433 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Wilson, D. N. Ribosome-targeting antibiotics and mechanisms of bacterial resistance. Nat. Rev. Microbiol. 12, 35–48 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Lin, J., Zhou, D., Steitz, T. A., Polikanov, Y. S. & Gagnon, M. G. Ribosome-targeting antibiotics: modes of action, mechanisms of resistance, and implications for drug design. Annu. Rev. Biochem. 87, 451–478 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shen, J., Wang, Y. & Schwarz, S. Presence and dissemination of the multiresistance gene cfr in Gram-positive and Gram-negative bacteria. J. Antimicrob. Chemother. 68, 1697–1706 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Vester, B. The cfr and cfr-like multiple resistance genes. Res. Microbiol. 169, 61–66 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Giessing, A. M. et al. Identification of 8-methyladenosine as the modification catalyzed by the radical SAM methyltransferase Cfr that confers antibiotic resistance in bacteria. RNA 15, 327–336 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Long, K. S., Poehlsgaard, J., Kehrenberg, C., Schwarz, S. & Vester, B. The Cfr rRNA methyltransferase confers resistance to Phenicols, Lincosamides, Oxazolidinones, Pleuromutilins, and Streptogramin A antibiotics. Antimicrob. Agents Chemother. 50, 2500–2505 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Smith, L. K. & Mankin, A. S. Transcriptional and translational control of the mlr operon, which confers resistance to seven classes of protein synthesis inhibitors. Antimicrob. Agents Chemother. 52, 1703–1712 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Polikanov, Y. S. et al. Distinct tRNA accommodation intermediates observed on the ribosome with the antibiotics hygromycin A and A201A. Mol. Cell 58, 832–844 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Toh, S. M., Xiong, L., Bae, T. & Mankin, A. S. The methyltransferase YfgB/RlmN is responsible for modification of adenosine 2503 in 23S rRNA. RNA 14, 98–106 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yan, F. et al. RlmN and Cfr are radical SAM enzymes involved in methylation of ribosomal RNA. J. Am. Chem. Soc. 132, 3953–3964 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Grove, T. L. et al. A radically different mechanism for S-adenosylmethionine-dependent methyltransferases. Science 332, 604–607 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Vazquez-Laslop, N., Ramu, H., Klepacki, D., Kannan, K. & Mankin, A. S. The key function of a conserved and modified rRNA residue in the ribosomal response to the nascent peptide. EMBO J. 29, 3108–3117 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ramu, H. et al. Nascent peptide in the ribosome exit tunnel affects functional properties of the A-site of the peptidyl transferase center. Mol. Cell 41, 321–330 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Benitez-Paez, 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  CAS  PubMed  PubMed Central  Google Scholar 

  16. Atkinson, G. C. et al. Distinction between the Cfr methyltransferase conferring antibiotic resistance and the housekeeping RlmN methyltransferase. Antimicrob. Agents Chemother. 57, 4019–4026 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Madhaiyan, M., Wirth, J. S. & Saravanan, V. S. Phylogenomic analyses of the Staphylococcaceae family suggest the reclassification of five species within the genus Staphylococcus as heterotypic synonyms, the promotion of five subspecies to novel species, the taxonomic reassignment of five Staphylococcus species to Mammaliicoccus gen. nov., and the formal assignment of Nosocomiicoccus to the family Staphylococcaceae. Int. J. Syst. Evol. Microbiol. 70, 5926–5936 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Schwarz, S., Werckenthin, C. & Kehrenberg, C. Identification of a plasmid-borne chloramphenicol-florfenicol resistance gene in Staphylococcus sciuri. Antimicrob. Agents Chemother. 44, 2530–2533 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Toh, S. M. et al. Acquisition of a natural resistance gene renders a clinical strain of methicillin-resistant Staphylococcus aureus resistant to the synthetic antibiotic linezolid. Mol. Microbiol. 64, 1506–1514 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hansen, L. H., Planellas, M. H., Long, K. S. & Vester, B. The order Bacillales hosts functional homologs of the worrisome cfr antibiotic resistance gene. Antimicrob. Agents Chemother. 56, 3563–3567 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tsai, K. et al. Directed evolution of the rRNA methylating enzyme Cfr reveals molecular basis of antibiotic resistance. eLife 11, e70017 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schmeing, T. M., Huang, K. S., Strobel, S. A. & Steitz, T. A. An induced-fit mechanism to promote peptide bond formation and exclude hydrolysis of peptidyl-tRNA. Nature 438, 520–524 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Polikanov, Y. S., Steitz, T. A. & Innis, C. A. A proton wire to couple aminoacyl-tRNA accommodation and peptide-bond formation on the ribosome. Nat. Struct. Mol. Biol. 21, 787–793 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Svetlov, M. S. et al. Structure of Erm-modified 70S ribosome reveals the mechanism of macrolide resistance. Nat. Chem. Biol. 17, 412–420 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Syroegin, E. A., Aleksandrova, E. V. & Polikanov, Y. S. Structural basis for the inability of chloramphenicol to inhibit peptide bond formation in the presence of A-site glycine. Nucleic Acids Res. 50, 7669–7679 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Syroegin, E. A., Aleksandrova, E. V. & Polikanov, Y. S. Insights into the ribosome function from the structures of non-arrested ribosome-nascent chain complexes. Nat. Chem. 15, 143–153 (2023).

    Article  CAS  PubMed  Google Scholar 

  27. Polikanov, Y. S., Melnikov, S. V., Soll, D. & Steitz, T. A. Structural insights into the role of rRNA modifications in protein synthesis and ribosome assembly. Nat. Struct. Mol. Biol. 22, 342–344 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tsai, K. et al. Structural basis for context-specific inhibition of translation by oxazolidinone antibiotics. Nat. Struct. Mol. Biol. 29, 162–171 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Syroegin, E. A. et al. Structural basis for the context-specific action of the classic peptidyl transferase inhibitor chloramphenicol. Nat. Struct. Mol. Biol. 29, 152–161 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Svetlov, M. S. et al. High-resolution crystal structures of ribosome-bound chloramphenicol and erythromycin provide the ultimate basis for their competition. RNA 25, 600–606 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tereshchenkov, A. G. et al. Binding and action of amino acid analogs of chloramphenicol upon the bacterial ribosome. J. Mol. Biol. 430, 842–852 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chen, C. W. et al. Binding and action of triphenylphosphonium analog of chloramphenicol upon the bacterial ribosome. Antibiotics 10, 390 (2021).

  33. Asai, T., Zaporojets, D., Squires, C. & Squires, C. L. An Escherichia coli strain with all chromosomal rRNA operons inactivated: complete exchange of rRNA genes between bacteria. Proc. Natl Acad. Sci. USA 96, 1971–1976 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hansen, J. L. et al. The structures of four macrolide antibiotics bound to the large ribosomal subunit. Mol. Cell 10, 117–128 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Wu, K. J. Y., Klepacki, D., Mankin, A. S. & Myers, A. G. A method for tritiation of iboxamycin permits measurement of its ribosomal binding. Bioorg. Med. Chem. Lett. 91, 129364 (2023).

    Article  CAS  PubMed  Google Scholar 

  36. Mitcheltree, M. J. et al. A synthetic antibiotic class overcoming bacterial multidrug resistance. Nature 599, 507–512 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Brodiazhenko, T. et al. Synthetic oxepanoprolinamide iboxamycin is active against Listeria monocytogenes despite the intrinsic resistance mediated by VgaL/Lmo0919 ABCF ATPase. JAC Antimicrob. Resist. 4, dlac061 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Obana, N. et al. Genome-encoded ABCF factors implicated in intrinsic antibiotic resistance in Gram-positive bacteria: VmlR2, Ard1 and CplR. Nucleic Acids Res. 51, 4536–4554 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pfister, P. et al. The structural basis of macrolide-ribosome binding assessed using mutagenesis of 23S rRNA positions 2058 and 2059. J. Mol. Biol. 342, 1569–1581 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Liu, M. & Douthwaite, S. Resistance to the macrolide antibiotic tylosin is conferred by single methylations at 23S rRNA nucleotides G748 and A2058 acting in synergy. Proc. Natl Acad. Sci. USA 99, 14658–14663 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li, Q. et al. Synthetic group A streptogramin antibiotics that overcome Vat resistance. Nature 586, 145–150 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Goethe, O., DiBello, M. & Herzon, S. B. Total synthesis of structurally diverse pleuromutilin antibiotics. Nat. Chem. 14, 1270–1277 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Osterman, I. A. et al. Madumycin II inhibits peptide bond formation by forcing the peptidyl transferase center into an inactive state. Nucleic Acids Res. 45, 7507–7514 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Dunkle, J. A., Xiong, L., Mankin, A. S. & Cate, J. H. Structures of the Escherichia coli ribosome with antibiotics bound near the peptidyl transferase center explain spectra of drug action. Proc. Natl Acad. Sci. USA 107, 17152–17157 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mason, J. D., Terwilliger, D. W., Pote, A. R. & Myers, A. G. Practical gram-scale synthesis of iboxamycin, a potent antibiotic candidate. J. Am. Chem. Soc. 143, 11019–11025 (2021).

    Article  CAS  PubMed  Google Scholar 

  46. McArthur, A. G. et al. The Comprehensive Antibiotic Resistance Database. Antimicrob. Agents Chemother. 57, 3348–3357 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Katoh, K. & Standley, D. M. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol. Biol. Evol. 30, 772–780 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Capella-Gutierrez, S., Silla-Martinez, J. M. & Gabaldon, T. trimAl: a tool for automated alignment trimming in large-scale phylogenetic analyses. Bioinformatics 25, 1972–1973 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Price, M. N., Dehal, P. S. & Arkin, A. P. FastTree 2—approximately maximum-likelihood trees for large alignments. PLoS ONE 5, e9490 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  50. UniProt, C. Uniprot The universal protein knowledgebase in 2023. Nucleic Acids Res. 51, D523–D531 (2023).

    Article  Google Scholar 

  51. Nguyen, L. T., Schmidt, H. A., von Haeseler, A. & Minh, B. Q. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol. Biol. Evol. 32, 268–274 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Miller, M. A. et al. A RESTful API for access to phylogenetic tools via the CIPRES science gateway. Evol. Bioinform. Online 11, 43–48 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Carr, J. F., Danziger, M. E., Huang, A. L., Dahlberg, A. E. & Gregory, S. T. Engineering the genome of Thermus thermophilus using a counterselectable marker. J. Bacteriol. 197, 1135–1144 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nakamura, A., Takakura, Y., Kobayashi, H. & Hoshino, T. In vivo directed evolution for thermostabilization of Escherichia coli hygromycin B phosphotransferase and the use of the gene as a selection marker in the host-vector system of Thermus thermophilus. J. Biosci. Bioeng. 100, 158–163 (2005).

    Article  CAS  PubMed  Google Scholar 

  55. Hashimoto, Y., Yano, T., Kuramitsu, S. & Kagamiyama, H. Disruption of Thermus thermophilus genes by homologous recombination using a thermostable kanamycin-resistant marker. FEBS Lett. 506, 231–234 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Douthwaite, S., Powers, T., Lee, J. Y. & Noller, H. F. Defining the structural requirements for a helix in 23S ribosomal RNA that confers erythromycin resistance. J. Mol. Biol. 209, 655–665 (1989).

    Article  CAS  PubMed  Google Scholar 

  57. Bollenbach, T., Quan, S., Chait, R. & Kishony, R. Nonoptimal microbial response to antibiotics underlies suppressive drug interactions. Cell 139, 707–718 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kannan, K., Vazquez-Laslop, N. & Mankin, A. S. Selective protein synthesis by ribosomes with a drug-obstructed exit tunnel. Cell 151, 508–520 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Quan, S., Skovgaard, O., McLaughlin, R. E., Buurman, E. T. & Squires, C. L. Markerless Escherichia coli rrn deletion strains for genetic determination of ribosomal binding sites. G3 5, 2555–2557 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Gamper, H. & Hou, Y.M. tRNA 3′-amino-tailing for stable amino acid attachment. RNA 24, 1878–1885 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kabsch, W. XDS. Acta Crystallogr. D 66, 125–132 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. McCoy, A. J. et al. PHASER crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Mankin and N. Vazquez-Laslop for providing E. coli SQ171 ΔtolC strains and for valuable discussions. We thank the staff at NE-CAT beamlines 24ID-C and 24ID-E for help with X-ray diffraction data collection, especially M. Capel, F. Murphy, S. Banerjee, I. Kourinov, D. Neau, J. Schuermann, N. Sukumar, A. Lynch, J. Withrow, K. Perry, A. Kaya and C. Salbego. This work is based upon research conducted at the Northeastern Collaborative Access Team beamlines, which are funded by the National Institute of General Medical Sciences from the National Institutes of Health (NIH) (grant on. P30-GM124165 to NE-CAT). The Eiger 16M detector on 24ID-E beamline is funded by an NIH-ORIP HEI grant (grant no. S10-OD021527 to NE-CAT). This research used resources of the Advanced Photon Source, a US Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under contract no. DE-AC02-06CH11357. This work was supported by the National Institute of Allergy and Infectious Diseases of the NIH (grant no. R01-AI168228 to A.G.M. and grant no. R21-AI163466 to Y.S.P.), the National Institute of General Medical Sciences of the NIH (grant no. R01-GM094157 to S.T.G. and grant no. R01-GM132302 to Y.S.P.), the National Science Foundation (grant no. MCB-1907273 to Y.S.P.), the USDA National Institute for Food and Agriculture (Hatch Project no. 1016013 to S.T.G.), the Illinois State startup funds (to Y.S.P.), the Swedish Research Council (Ventenskapsrådet) (grant nos. 2019-01085 and 2022-01603 to G.C.A.), the Knut and Alice Wallenberg Foundation (grant no. 2020.0037 to G.C.A.) and the Carl Tryggers Stiftelse för Vetenskaplig Forskning (grant no. CTS19:24 to G.C.A.). K.J.Y.W. was supported by a National Science Scholarship (Ph.D.) from the Agency for Science, Technology and Research (Singapore). The funders had no role in study design, data collection and analysis, decision to publish or manuscript preparation.

Author information

Authors and Affiliations

Authors

Contributions

E.V.A. with help from S.M.B. and M.S.S. constructed the T. thermophilus HB27 strain expressing Cfr-like methylases. G.C.A. performed phylogenetic analysis and identified putative thermostable cfr-like genes. B.I.C.T. and K.J.Y.W. synthesized iboxamycin. E.E.K. prepared ΔrlmN knock-out T. thermophilus HB27 strain. E.V.A. performed the assessment of A2503-C8-methylation using primer extension assay. E.V.A. and E.A.S. grew T. thermophilus cells and purified Cfr-modified 70S ribosomes. E.A.S. and E.V.A. prepared hydrolysis-resistant aminoacyl- and peptidyl-tRNAs. E.V.A. with help from B.I.C.T. and K.J.Y.W. performed microbiological assays. E.V.A., E.A.S. and Y.S.P. designed and performed X-ray crystallography experiments. A.G.M., Y.S.P. and S.T.G. supervised the experiments. All authors interpreted the results. E.V.A., B.I.C.T., K.J.Y.W., A.G.M. and Y.S.P. wrote the manuscript.

Corresponding authors

Correspondence to Andrew G. Myers or Yury S. Polikanov.

Ethics declarations

Competing interests

A.G.M. is an inventor in a provisional patent application submitted by the President and Fellows of Harvard College covering oxepanoprolinamide antibiotics described in this work. A.G.M. has filed the following international patent applications: WO/2019/032936 ‘Lincosamide Antibiotics and Uses Thereof’ and WO/2019/032956 ‘Lincosamide Antibiotics and Uses Thereof’. The other authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Comparison of the structures of Cfr-modified and wild-type 70S ribosomes from T. thermophilus and E. coli.

Superpositioning of the previously reported structures of T. thermophilus WT 70S ribosome containing aminoacylated Phe-tRNAPhe in the A site and either fMet-tRNAiMet (a, b; PDB entry 6XHW ref. 24) or fMTHSMRC-peptidyl tRNAiMet (c, d; PDB entry 8CVL ref. 26) in the P site, or E. coli WT 70S ribosome containing radezolid in the A site and fMFKAF-peptidyl-tRNAPhe in the P site (e, f; PDB entry 7S1I ref. 28) with the structures of the same complexes containing Cfr-modified nucleotide A2503 of the 23S rRNA. All structures were aligned based on domain V of the 23S rRNA. (a, c, e) Comparisons of the positions of key 23S rRNA nucleotides around the PTC. (b, d, f) Comparisons of the positions of A- and P-site substrates relative to nucleotides A2062 and A2503. Nucleotides of the Cfr-modified and unmodified ribosomes are shown in blue and light blue, espectively. The Cfr-modified residue A2503 is highlighted in navy blue, with the C8-methyl group shown in orange. E. coli nucleotide numbering is used. H-bonds are shown with dotted lines.

Extended Data Fig. 2 Schematic diagrams of H-bond rearrangement between nucleotides in position 2062 and A2503 of the 23S rRNA upon Hoogsteen base pair formation.

(a) Formation of the symmetric trans A-A Hoogsteen base pair between A2062 and m2A2503 observed in the structures of 70S ribosome. Note that the formation of this base pair requires the N7-atoms of both adenines to be deprotonated in order to serve as H-bond acceptors of the N6-protons of the base-paired nucleotide. (b) The same Hoogsteen base pair is impossible with a guanine nucleotide in position 2062 of the 23S rRNA due to the inability to form an H-bond between O6 of G2062 and N7 of m2A2503.

Extended Data Fig. 3 Electron density maps of 23S rRNA in wild-type and Cfr-modified T. thermophilus 70S ribosome.

2Fo-Fc electron difference Fourier maps (blue mesh) of A2062 and A2503 residues of 23S rRNA in the wild-type (a) or Cfr-modified (b, c) T. thermophilus 70S ribosome carrying aminoacylated Phe-tRNAPhe in the A site and either fMet-tRNAiMet (a, b) or fMTHSMRC-peptidyl-tRNAiMet (c) in the P site. The structure and the electron density map of the wild-type ribosome complex (a) are from PDB entry 6XHW ref. 24. Carbon atoms are colored light blue for the C8-unmethylated A2503 (a) and blue for the Cfr-modified A2503 (b, c); nitrogens are dark blue; oxygens are red.

Extended Data Fig. 4 Interactions of fMTHSMRC-peptidyl-tRNAs with wild-type and Cfr-modified T. thermophilus 70S ribosome.

Close-up views of the aminoacyl and peptidyl moieties of A-site Phe-tRNAPhe and P-site fMTHSMRC-tRNAiMet in the wild-type (a, b; PDB entry 8CVL ref. 26) or Cfr-modified (c, d) T. thermophilus 70S ribosome. H-bonds are shown by black dotted lines. Stacking interactions between the aromatic side chain of His3 of fMTHSMRC-peptidyl-tRNA and A2062 nucleobase of the 23S rRNA are indicated by a black arrow.

Extended Data Fig. 5 Comparison of the structures of WT and Cfr-modified ribosomes from E. coli and T. thermophilus.

(a) Superpositioning of the previous structures of WT 70S ribosomes carrying P-site peptidyl-tRNAs from T. thermophilus (light blue, PDB entry 8CVL ref. 26) and E. coli (teal, PDB entry 7S1I ref. 28). (b) Superpositioning of the new structure of Cfr-modified 70S ribosome carrying P-site peptidyl-tRNAs from T. thermophilus (blue) and E. coli (light teal, PDB entry 7S1K ref. 28).

Extended Data Fig. 6 Comparison of electron density maps of iboxamycin (IBX) in complex with Cfr-modified and wild-type T. thermophilus 70S ribosomes.

2Fo-Fc electron density maps (blue mesh) contoured at 1.0σ of IBX in complex with Cfr-modified (a, b, yellow) or wild-type (c, d, teal) T. thermophilus 70S ribosomes. The C8-methyl group of m2m8A2503 is highlighted in orange. The structure and the electron density map of IBX in complex with wild-type 70S ribosome (c, d) are from PDB entry 7RQ8 ref. 36. Carbon atoms are colored navy blue for the Cfr-modified m2m8A2503 (a, b) and light blue for the WT m2A2503 (c, d); nitrogens are dark blue; oxygens are red.

Extended Data Fig. 7 Comparison of the m2m8A2503 positions in the Cfr-modified ribosome in the presence and absence of iboxamycin.

(a) Superposition of the structures of drug-free Cfr-modified 70S ribosome containing m2m8A2503 residue (shown as navy blue spheres with C8-methyl group highlighted in red) with the structure of ribosome-bound iboxamycin (IBX, yellow). (b) Structure of Cfr-modified 70S ribosome containing m2m8A2503 residue (shown as blue spheres with C8-methyl highlighted in orange) in complex with iboxamycin (IBX, yellow). Note that binding of iboxamycin to the Cfr-modified ribosome causes an ~1 Å shift of the m2m8A2503 residue away from the drug.

Extended Data Fig. 8 Structural basis for the Cfr-mediated resistance to 16-membered macrolides.

(a–c) Chemical structures of tylosin (a), spiramycin (b), and carbomycin (c). (d-f) Superposition of the structures of Cfr-modified T. thermophilus 70S ribosome containing C8-methylated A2503 residue in the 23S rRNA (blue) with the previously reported structures of 16-membered macrolides, such as tylosin (d, green; PDB entry 1K9M ref. 34), spiramycin (e, light teal; PDB entry 1KD1 ref. 34), or carbomycin (f, tealF PDB entry 1K8A ref. 34) in complex with the 50S ribosomal subunit from the archaeon H. marismortui. The degrees of steric overlaps between the C8-methyl group of the m2m8A2503 nucleotide and each PTC-acting drug are shown in yellow. These numbers reflect the distance in Å that the drug and the m2m8A2503 residue need to move away from each other to avoid the steric clash. Note that the C8-methyl group of m2m8A2503 (highlighted in orange) can physically interfere with the binding of 16-membered macrolides.

Extended Data Fig. 9 Structures of tylosin (TYL) bound to the Cfr-modified and WT 70S ribosomes.

(a–f) Electron density map (blue mesh) contoured at 1.0σ of TYL (green or magenta) in complex with the Cfr-modified (a–c) or wild-type (d–f) T. thermophilus 70S ribosome containing m2m8A2503 (dark blue with C8-methyl group highlighted in orange) or m2A2503 (light blue) residues in the 23S rRNA, respectively. The chemical structure of tylosin is shown in panel d. The reactive acetaldehyde group (highlighted in red) at C6 of tylosin’s macrolactone ring forms a covalent bond with the exocyclic N6-amino group of A2062 in the T. thermophilus ribosome. Note that the mycinose moiety at C14 of the macrolactone ring of tylosin is well-resolved in the electron density maps.

Extended Data Fig. 10 Comparisons of A2503 positions in Cfr-modified ribosomes in the presence and absence of iboxamycin or tylosin.

Superposition of the structures of Cfr-modified 70S ribosome both containing m2m8A2503 residue in the presence (blue) and absence (navy blue) of iboxamycin (a, b, yellow) or tylosin (c, d, green). Note that while binding of iboxamycin to the Cfr-modified ribosome causes an ~1 Å shift of m2m8A2503 residue away from the drug, binding of tylosin does not affect the position of the m2m8A2503 residue.

Supplementary information

Supplementary Information

Supplementary Tables 1–5, Figs. 1 and 2 and references.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed gel for Fig. 1c.

Source Data Fig. 2

Unprocessed gel for Fig. 1d.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aleksandrova, E.V., Wu, K.J.Y., Tresco, B.I.C. et al. Structural basis of Cfr-mediated antimicrobial resistance and mechanisms to evade it. Nat Chem Biol (2024). https://doi.org/10.1038/s41589-023-01525-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-023-01525-w

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