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.

  • Review Article
  • Published:

Ribosome-targeting antibiotics and mechanisms of bacterial resistance

Key Points

  • The bacterial ribosome is one of the main targets of antibiotics, with most clinically used antibiotics targeting either the decoding site on the small ribosomal subunit (30S subunit) or the peptidyl-transferase centre on the large subunit (50S subunit).

  • The majority of antibiotics that target the 30S subunit inhibit protein synthesis by preventing either the binding of tRNAs to the ribosome or the movement of tRNAs through the ribosome during translocation.

  • Most antibiotics that target the 50S subunit inhibit protein synthesis by either perturbing the binding of aminoacylated-tRNAs at the A- or P-sites or preventing the channelling of the nascent polypeptide chain through the ribosomal tunnel.

  • Bacterial antibiotic resistance mechanisms include efflux, reduced influx, modification and degradation of the drug, as well as mutation, modification or overexpression of the target.

  • The majority of ribosome-targeting antibiotics in clinical trials are semi-synthetic derivatives of naturally produced compounds, but further work will be required to develop antibiotics that target novel sites on the ribosome.

Abstract

The ribosome is one of the main antibiotic targets in the bacterial cell. Crystal structures of naturally produced antibiotics and their semi-synthetic derivatives bound to ribosomal particles have provided unparalleled insight into their mechanisms of action, and they are also facilitating the design of more effective antibiotics for targeting multidrug-resistant bacteria. In this Review, I discuss the recent structural insights into the mechanism of action of ribosome-targeting antibiotics and the molecular mechanisms of bacterial resistance, in addition to the approaches that are being pursued for the production of improved drugs that inhibit bacterial protein synthesis.

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

Figure 1: Antibiotic target sites during bacterial protein synthesis.
Figure 2: Antibiotic binding sites on the 30S and 50S ribosomal subunits.
Figure 3: Antibiotic interactions at the decoding centre of the 30S subunit.
Figure 4: Antibiotic interactions at the peptidyl-transferase centre of the 50S subunit.
Figure 5: Antibacterial resistance mechanisms.

Similar content being viewed by others

References

  1. Voorhees, R. M. & Ramakrishnan, V. Structural basis of the translational elongation cycle. Annu. Rev. Biochem. 82, 203–236 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Walsh, C. Where will new antibiotics come from? Nature Rev. Microbiol. 1, 65–70 (2003).

    Article  CAS  Google Scholar 

  3. Poehlsgaard, J. & Douthwaite, S. The bacterial ribosome as a target for antibiotics. Nature Rev. Microbiol. 3, 870–881 (2005).

    Article  CAS  Google Scholar 

  4. Wilson, D. N. The A-Z of bacterial translation inhibitors. Crit. Rev. Biochem. Mol. Biol. 44, 393–433 (2009). A comprehensive overview of the mechanism of action of antibiotics that target the bacterial translation apparatus.

    Article  CAS  PubMed  Google Scholar 

  5. Sohmen, D., Harms, J. M., Schlunzen, F. & Wilson, D. N. SnapShot: Antibiotic inhibition of protein synthesis I. Cell 138, 1248 e1241 (2009).

    Article  PubMed  Google Scholar 

  6. Savelsbergh, A., Rodnina, M. V. & Wintermeyer, W. Distinct functions of elongation factor G in ribosome recycling and translocation. RNA 15, 772–780 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Svidritskiy, E., Ling, C., Ermolenko, D. N. & Korostelev, A. A. Blasticidin S inhibits translation by trapping deformed tRNA on the ribosome. Proc. Natl Acad. Sci. USA 110, 12283–12288 (2013).

    Article  PubMed  Google Scholar 

  8. Selmer, M. et al. Structure of the 70S ribosome complexed with mRNA and tRNA. Science 313, 1935–1942 (2006). The high-resolution crystal structure of the T. thermophilus 70S ribosome reported in this study opened the way to structures of T. thermophilus 70S–antibiotic complexes, such as those reported in references 7, 18, 29, 30 and 40.

    Article  CAS  PubMed  Google Scholar 

  9. Schuwirth, B. et al. Structures of the bacterial ribosome at 3.5 Å resolution. Science 310, 827–834 (2005). The high-resolution crystal structure of the E. coli 70S ribosome reported here opened the way to structures of E. coli 70S–antibiotic complexes, such as those reported in references 24, 25, 41, 139 and 141.

    Article  CAS  PubMed  Google Scholar 

  10. Belova, L., Tenson, T., Xiong, L. Q., McNicholas, P. M. & Mankin, A. S. A novel site of antibiotic action in the ribosome: interaction of evernimicin with the large ribosomal subunit. Proc. Natl Acad. Sci. USA 98, 3726–3731 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Mikolajka, A. et al. Differential effects of thiopeptide and orthosomycin antibiotics on translational GTPases. Chem. Biol. 18, 589–600 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fischbach, M. A. & Walsh, C. T. Antibiotics for emerging pathogens. Science 325, 1089–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Brodersen, D. E. et al. The structural basis for the action of the antibiotics tetracycline, pactamycin, and hygromycin B on the 30S ribosomal subunit. Cell 103, 1143–1154 (2000). This study, together with references 14, 20 and 142, reports the first structures of antibiotics in complex with the 30S subunit.

    Article  CAS  PubMed  Google Scholar 

  14. Pioletti, M. et al. Crystal structures of complexes of the small ribosomal subunit with tetracycline, edeine and IF3. EMBO J. 20, 1829–1839 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Roberts, M. C. Update on acquired tetracycline resistance genes. FEMS Microbiol. Lett. 245, 195–203 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Olson, M. W. et al. Functional, biophysical, and structural bases for antibacterial activity of tigecycline. Antimicrob. Agents Chemother. 50, 2156–2166 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Grossman, T. H. et al. Target- and resistance-based mechanistic studies with TP-434, a novel fluorocycline antibiotic. Antimicrob. Agents Chemother. 56, 2559–2564 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jenner, L. et al. Structural basis for potent inhibitory activity of the antibiotic tigecycline during protein synthesis. Proc. Natl Acad. Sci. USA 110, 3812–3816 (2013).

    Article  PubMed  Google Scholar 

  19. Fourmy, D., Recht, M. I., Blanchard, S. C. & Puglisi, J. D. Structure of the A-site of E. coli 16S ribosomal RNA complexed with an aminoglycoside antibiotic. Science 274, 1367–1371 (1996).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  21. Ogle, J. M. & Ramakrishnan, V. Structural insights into translational fidelity. Annu. Rev. Biochem. 74, 129–177 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Demeshkina, N., Jenner, L., Westhof, E., Yusupov, M. & Yusupova, G. A new understanding of the decoding principle on the ribosome. Nature 484, 256–259 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Matt, T. et al. Dissociation of antibacterial activity and aminoglycoside ototoxicity in the 4-monosubstituted 2-deoxystreptamine apramycin. Proc. Natl Acad. Sci. USA 109, 10984–10989 (2012).

    Article  PubMed  Google Scholar 

  24. Borovinskaya, M. A. et al. Structural basis for aminoglycoside inhibition of bacterial ribosome recycling. Nature Struct. Mol. Biol. 14, 727–732 (2007).

    Article  CAS  Google Scholar 

  25. Wang, L. et al. Allosteric control of the ribosome by small-molecule antibiotics. Nature Struct. Mol. Biol. 19, 957–963 (2012).

    Article  CAS  Google Scholar 

  26. Cabanas, M. J., Vazquez, D. & Modolell, J. Inhibition of ribosomal translocation by aminoglycoside antibiotics. Biochem. Biophys. Res. Commun. 83, 991–997 (1978).

    Article  CAS  PubMed  Google Scholar 

  27. Misumi, M., Nishimura, T., Komai, T. & Tanaka, N. Interaction of kanamycin and related antibiotics with the large subunit of ribosomes and the inhibition of translocation. Biochem. Biophys. Res. Commun. 84, 358–365 (1978).

    Article  CAS  PubMed  Google Scholar 

  28. Feldman, M. B., Terry, D. S., Altman, R. B. & Blanchard, S. C. Aminoglycoside activity observed on single pre-translocation ribosome complexes. Nature Chem. Biol. 6, 54–62 (2010).

    Article  CAS  Google Scholar 

  29. Stanley, R. E., Blaha, G., Grodzicki, R. L., Strickler, M. D. & Steitz, T. A. The structures of the anti-tuberculosis antibiotics viomycin and capreomycin bound to the 70S ribosome. Nature Struct. Mol. Biol. 17, 289–293 (2010).

    Article  CAS  Google Scholar 

  30. Bulkley, D., Johnson, F. & Steitz, T. A. The antibiotic thermorubin inhibits protein synthesis by binding to inter-subunit bridge B2a of the ribosome. J. Mol. Biol. 416, 571–578 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jain, A. & Dixit, P. Multidrug-resistant to extensively drug resistant tuberculosis: what is next? J. Biosci. 33, 605–616 (2008).

    Article  PubMed  Google Scholar 

  32. Modolell, J. & Vazquez, D. The inhibition of ribosomal translocation by viomycin. Eur. J. Biochem. 81, 491–497 (1977).

    Article  CAS  PubMed  Google Scholar 

  33. Ermolenko, D. N. et al. The antibiotic viomycin traps the ribosome in an intermediate state of translocation. Nature Struct. Mol. Biol. 14, 493–497 (2007).

    Article  CAS  Google Scholar 

  34. Cornish, P. V., Ermolenko, D. N., Noller, H. F. & Ha, T. Spontaneous intersubunit rotation in single ribosomes. Mol. Cell 30, 578–588 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Akbergenov, R. et al. Molecular basis for the selectivity of antituberculosis compounds capreomycin and viomycin. Antimicrob. Agents Chemother. 55, 4712–4717 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cavalleri, B., Turconi, M. & Pallanza, R. Synthesis and antibacterial activity of some derivatives of the antibiotic thermorubin. J. Antibiot. (Tokyo) 38, 1752–1760 (1985).

    Article  CAS  Google Scholar 

  37. Lin, F. & Wishnia, A. The protein synthesis inhibitor thermorubin. 1. Nature of the thermorubin-ribosome complex. Biochemistry 21, 477–483 (1982).

    Article  CAS  PubMed  Google Scholar 

  38. Lin, F. & Wishnia, A. The protein synthesis inhibitor thermorubin. 2. Mechanism of inhibition of initiation on Escherichia coli ribosomes. Biochemistry 21, 484–491 (1982).

    Article  CAS  PubMed  Google Scholar 

  39. Wilson, D. N. On the specificity of antibiotics targeting the large ribosomal subunit. Ann. NY Acad. Sci. 1241, 1–16 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Bulkley, D., Innis, C. A., Blaha, G. & Steitz, T. A. Revisiting the structures of several antibiotics bound to the bacterial ribosome. Proc. Natl Acad. Sci. USA 107, 17158–17163 (2010).

    Article  PubMed  Google Scholar 

  41. 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). References 40 and 41 reveal that macrolides bind to bacterial ribosomes, as reported previously for the archaeal 50S subunit. However, the data obtained question the validity of the conformation of macrolides and chloramphenicol reported in the earlier bacterial D. radiodurans structures (for example, reference 46).

    Article  PubMed  Google Scholar 

  42. Blaha, G. M., Polikanov, Y. S. & Steitz, T. A. Elements of ribosomal drug resistance and specificity. Curr. Opin. Struct. Biol. 22, 750–758 (2013).

    Article  CAS  Google Scholar 

  43. Hansen, J. L. et al. The structures of four macrolide antibiotics bound to the large ribosomal subunit. Mol. Cell 10, 117–128 (2002). Together with references 46 and 47, this study reports the first structures of antibiotics in complex with the 50S subunit.

    Article  CAS  PubMed  Google Scholar 

  44. Tu, D., Blaha, G., Moore, P. & Steitz, T. Structures of MLSBK antibiotics bound to mutated large ribosomal subunits provide a structural explanation for resistance. Cell 121, 257–270 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Gurel, G., Blaha, G., Steitz, T. A. & Moore, P. B. Structures of triacetyloleandomycin and mycalamide A bind to the large ribosomal subunit of Haloarcula marismortui. Antimicrob. Agents Chemother. 53, 5010–5014 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schlünzen, F. et al. Structural basis for the interaction of antibiotics with the peptidyl transferase centre in eubacteria. Nature 413, 814–821 (2001).

    Article  PubMed  Google Scholar 

  47. Hansen, J. L., Moore, P. B. & Steitz, T. A. Structures of five antibiotics bound at the peptidyl transferase center of the large ribosomal subunit. J. Mol. Biol. 330, 1061–1075 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Mankin, A. S. & Garrett, R. A. Chloramphenicol resistance mutations in the single 23S rRNA gene of the archaeon Halobacterium halobium. J. Bacteriol. 173, 3559–3563 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Champney, W. S. The other target for ribosomal antibiotics: inhibition of bacterial ribosomal subunit formation. Infect. Disord. Drug Targets 6, 377–390 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Siibak, T. et al. Antibiotic-induced ribosomal assembly defects result from changes in the synthesis of ribosomal proteins. Mol. Microbiol. 80, 54–67 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Siibak, T. et al. Erythromycin- and chloramphenicol-induced ribosomal assembly defects are secondary effects of protein synthesis inhibition. Antimicrob. Agents Chemother. 53, 563–571 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Leach, K. L. et al. The site of action of oxazolidinone antibiotics in living bacteria and in human mitochondria. Mol. Cell 26, 393–402 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Wilson, D. N. et al. The oxazolidinone antibiotics perturb the ribosomal peptidyl-transferase center and effect tRNA positioning. Proc. Natl Acad. Sci. USA 105, 13339–13344 (2008).

    Article  PubMed  Google Scholar 

  54. Ippolito, J. A. et al. Crystal structure of the oxazolidinone antibiotic linezolid bound to the 50S ribosomal subunit. J. Med. Chem. 51, 3353–3356 (2008). References 52–54 show excellent agreement for the binding position of the oxazolidinone linezolid at the PTC of bacterial and archaeal ribosomes.

    Article  CAS  PubMed  Google Scholar 

  55. Fredrick, K. & Noller, H. F. Catalysis of ribosomal translocation by sparsomycin. Science 300, 1159–1162 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Leach, K. L., Brickner, S. J., Noe, M. C. & Miller, P. F. Linezolid, the first oxazolidinone antibacterial agent. Ann. NY Acad. Sci. 1222, 49–54 (2011).

    Article  CAS  PubMed  Google Scholar 

  57. Franceschi, F. & Duffy, E. M. Structure-based drug design meets the ribosome. Biochem. Pharmacol. 71, 1016–1025 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Li, S., Cheng, X., Zhou, Y. & Xi, Z. Sparsomycin-linezolid conjugates can promote ribosomal translocation. Chembiochem 12, 2801–2806 (2011).

    Article  CAS  PubMed  Google Scholar 

  59. Ermolenko, D. N., Cornish, P. V., Ha, T. & Noller, H. F. Antibiotics that bind to the A site of the large ribosomal subunit can induce mRNA translocation. RNA 19, 158–166 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kannan, K. & Mankin, A. S. Macrolide antibiotics in the ribosome exit tunnel: species-specific binding and action. Ann. NY Acad. Sci. 1241, 33–47 (2012).

    Article  CAS  Google Scholar 

  61. Berisio, R. et al. Structural insight into the antibiotic action of telithromycin on resistant mutants. J. Bact. 185, 4276–4279 (2003).

    Article  CAS  PubMed  Google Scholar 

  62. Wilson, D. N., Harms, J. M., Nierhaus, K. H., Schlünzen, F. & Fucini, P. Species-specific antibiotic-ribosome interactions: Implications for drug development. Biol. Chem. 386, 1239–1252 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Tenson, T., Lovmar, M. & Ehrenberg, M. The mechanism of action of macrolides, lincosamides and streptogramin B reveals the nascent peptide exit path in the ribosome. J. Mol. Biol. 330, 1005–1014 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Tenson, T., Xiong, L. Q., Kloss, P. & Mankin, A. S. Erythromycin resistance peptides selected from random peptide libraries. J. Biol. Chem. 272, 17425–17430 (1997).

    Article  CAS  PubMed  Google Scholar 

  65. Ramu, H., Mankin, A. & Vazquez-Laslop, N. Programmed drug-dependent ribosome stalling. Mol. Microbiol. 71, 811–824 (2009).

    Article  CAS  PubMed  Google Scholar 

  66. Kannan, K., Vazquez-Laslop, N. & Mankin, A. S. Selective protein synthesis by ribosomes with a drug-obstructed exit tunnel. Cell 151, 508–520 (2012). This work demonstrates that macrolide antibiotics allow the synthesis of a small subset of proteins, thus conceptually changing our mechanistic understanding of the mechanism of action of this clinically important class of antibiotics.

    Article  CAS  PubMed  Google Scholar 

  67. Vannuffel, P. & Cocito, C. Mechanism of action of streptogramins and macrolides. Drugs 51, 20–30 (1996).

    Article  CAS  PubMed  Google Scholar 

  68. Harms, J., Schluenzen, F., Fucini, P., Bartels, H. & Yonath, A. Alterations at the peptidyl transferase centre of the ribosome induced by the synergistic action of the streptogramins dalfopristin and quinupristin. BMC Biol. 2, 4 (2004). Together with references 44 and 69–70, this study provides a structural basis for the synergistic activity of the streptogramin class of antibiotics.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Auerbach, T. et al. The structure of ribosome-lankacidin complex reveals ribosomal sites for synergistic antibiotics. Proc. Natl Acad. Sci. USA 107, 1983–1988 (2010).

    Article  PubMed  Google Scholar 

  70. Belousoff, M. J. et al. Crystal structure of the synergistic antibiotic pair, lankamycin and lankacidin, in complex with the large ribosomal subunit. Proc. Natl Acad. Sci. USA 108, 2717–2122 (2011).

    Article  PubMed  Google Scholar 

  71. D'Costa, V. M. et al. Antibiotic resistance is ancient. Nature 477, 457–461 (2011).

    Article  CAS  PubMed  Google Scholar 

  72. Davies, J. & Davies, D. Origins and evolution of antibiotic resistance. Microbiol. Mol. Biol. Rev. 74, 417–433 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Forsberg, K. J. et al. The shared antibiotic resistome of soil bacteria and human pathogens. Science 337, 1107–1111 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Allen, H. K. et al. Call of the wild: antibiotic resistance genes in natural environments. Nature Rev. Microbiol. 8, 251–259 (2010).

    Article  CAS  Google Scholar 

  75. He, X. et al. Structure of a cation-bound multidrug and toxic compound extrusion transporter. Nature 467, 991–994 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lu, M. et al. Structures of a Na+-coupled, substrate-bound MATE multidrug transporter. Proc. Natl Acad. Sci. USA 110, 2099–2104 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Tanaka, Y. et al. Structural basis for the drug extrusion mechanism by a MATE multidrug transporter. Nature 496, 247–251 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. van Veen, H. W. Structural biology: last of the multidrug transporters. Nature 467, 926–927 (2010).

    Article  CAS  PubMed  Google Scholar 

  79. Piddock, L. J. Multidrug-resistance efflux pumps — not just for resistance. Nature Rev. Microbiol. 4, 629–636 (2006).

    Article  CAS  Google Scholar 

  80. Davidovich, C. et al. Induced-fit tightens pleuromutilins binding to ribosomes and remote interactions enable their selectivity. Proc. Natl Acad. Sci. USA 104, 4291–4296 (2007).

    Article  CAS  PubMed  Google Scholar 

  81. Harms, J. M. et al. Translational regulation via L11: molecular switches on the ribosome turned on and off by thiostrepton and micrococcin. Mol. Cell 30, 26–38 (2008).

    Article  CAS  PubMed  Google Scholar 

  82. LaMarre, J. et al. The genetic environment of the cfr gene and the presence of other mechanisms account for the very high linezolid resistance of Staphylococcus epidermidis isolate 426-3147L. Antimicrob. Agents Chemother. 57, 1173–1179 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Billal, D. S., Feng, J., Leprohon, P., Legare, D. & Ouellette, M. Whole genome analysis of linezolid resistance in Streptococcus pneumoniae reveals resistance and compensatory mutations. BMC Genomics 12, 512 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Beauclerk, A. A. D. & Cundliffe, E. Sites of action of two ribosomal RNA methylases responsible for resistance to aminoglycosides. J. Mol. Biol. 193, 661–671 (1987).

    Article  CAS  PubMed  Google Scholar 

  85. Cundliffe, E. How antibiotic-producing organisms avoid suicide. Ann. Rev. Microbiol. 43, 207–233 (1989).

    Article  CAS  Google Scholar 

  86. 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 

  87. 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 

  88. 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 

  89. LaMarre, J. M., Locke, J. B., Shaw, K. J. & Mankin, A. S. Low fitness cost of the multidrug resistance gene cfr. Antimicrob. Agents Chemother. 55, 3714–3719 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Gupta, P., Sothiselvam, S., Vazquez-Laslop, N. & Mankin, A. S. Deregulation of translation due to post-transcriptional modification of rRNA explains why erm genes are inducible. Nature Commun. 4, 1984 (2013).

    Article  CAS  Google Scholar 

  91. Monshupanee, T., Johansen, S. K., Dahlberg, A. E. & Douthwaite, S. Capreomycin susceptibility is increased by TlyA-directed 2′-O-methylation on both ribosomal subunits. Mol. Microbiol. 85, 1194–1203 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. LaMarre, J. M., Howden, B. P. & Mankin, A. S. Inactivation of the indigenous methyltransferase RlmN in Staphylococcus aureus increases linezolid resistance. Antimicrob. Agents Chemother. 55, 2989–2991 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Okamoto, S. et al. Loss of a conserved 7-methylguanosine modification in 16S rRNA confers low-level streptomycin resistance in bacteria. Mol. Microbiol. 63, 1096–1106 (2007).

    Article  CAS  PubMed  Google Scholar 

  94. Thom, G. & Prescott, C. D. The selection in vivo and characterization of an RNA recognition motif for spectinomycin. Bioorgan. Med. Chem. 5, 1081–1086 (1997).

    Article  CAS  Google Scholar 

  95. Gross, S. et al. Amythiamicin D and related thiopeptides as inhibitors of the bacterial elongation factor EF-Tu: modification of the amino acid at carbon atom C2 of ring C dramatically influences the activity. Eur. J. Biochem. http://dx.doi.org/10.1002/cmdc.201300323 (2013).

  96. Connell, S. R., Tracz, D. M., Nierhaus, K. H. & Taylor, D. E. Ribosomal protection proteins and their mechanism of tetracycline resistance. Antimicrob. Agents Chemother. 47, 3675–3681 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. O'Neill, A. J. & Chopra, I. Molecular basis of fusB-mediated resistance to fusidic acid in Staphylococcus aureus. Mol. Microbiol. 59, 664–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Dönhöfer, A. et al. Structural basis for TetM-mediated tetracycline resistance. Proc. Natl Acad. Sci. USA 109, 16900–16905 (2012).

    Article  PubMed  Google Scholar 

  99. Li, W. et al. Mechanism of tetracycline resistance by ribosomal protection protein Tet(O). Nature Commun. 4, 1477 (2013). References 98 and 99 report structures of ribosome protection proteins on the ribosome, revealing that these factors confer tetracycline resistance by directly dislodging tetracycline from its ribosomal binding site rather than by an indirect mechanism as previously proposed.

    Article  CAS  Google Scholar 

  100. Cox, G. et al. Ribosome clearance by FusB-type proteins mediates resistance to the antibiotic fusidic acid. Proc. Natl Acad. Sci. USA 109, 2102–2107 (2012).

    Article  PubMed  Google Scholar 

  101. Guo, X. et al. Structure and function of FusB: an elongation factor G-binding fusidic acid resistance protein active in ribosomal translocation and recycling. Open Biol. 2, 120016 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Wright, G. D. Bacterial resistance to antibiotics: enzymatic degradation and modification. Adv. Drug Deliv. Rev. 57, 1451–1470 (2005).

    Article  CAS  PubMed  Google Scholar 

  103. Ramirez, M. S. & Tolmasky, M. E. Aminoglycoside modifying enzymes. Drug Resist. Updat. 13, 151–171 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Mosher, R. H. et al. Inactivation of chloramphenicol by O-phosphorylation. A novel resistance mechanism in Streptomyces venezuelae ISP5230, a chloramphenicol producer. J. Biol. Chem. 270, 27000–27006 (1995).

    Article  CAS  PubMed  Google Scholar 

  105. Rajesh, T. et al. Phosphorylation of chloramphenicol by a recombinant protein Yhr2 from Streptomyces avermitilis MA4680. Bioorg. Med. Chem. Lett. 23, 3614–3619 (2013).

    Article  CAS  PubMed  Google Scholar 

  106. Dhote, V., Gupta, S. & Reynolds, K. A. An O-phosphotransferase catalyzes phosphorylation of hygromycin A in the antibiotic-producing organism Streptomyces hygroscopicus. Antimicrob. Agents Chemother. 52, 3580–3588 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Volkers, G., Palm, G. J., Weiss, M. S., Wright, G. D. & Hinrichs, W. Structural basis for a new tetracycline resistance mechanism relying on the TetX monooxygenase. FEBS Lett. 585, 1061–1066 (2011).

    Article  CAS  PubMed  Google Scholar 

  108. Moore, I. F., Hughes, D. W. & Wright, G. D. Tigecycline is modified by the flavin-dependent monooxygenase TetX. Biochemistry 44, 11829–11835 (2005).

    Article  CAS  PubMed  Google Scholar 

  109. Palmer, A. C., Angelino, E. & Kishony, R. Chemical decay of an antibiotic inverts selection for resistance. Nature Chem. Biol. 6, 105–107 (2010).

    Article  CAS  Google Scholar 

  110. Butler, M. S. & Cooper, M. A. Antibiotics in the clinical pipeline in 2011. J. Antibiot. (Tokyo) 64, 413–425 (2011).

    Article  CAS  Google Scholar 

  111. Sutcliffe, J. A. Antibiotics in development targeting protein synthesis. Ann. NY Acad. Sci. 1241, 122–152 (2011).

    Article  CAS  PubMed  Google Scholar 

  112. Pokrovskaya, V. & Baasov, T. Dual-acting hybrid antibiotics: a promising strategy to combat bacterial resistance. Expert Opin. Drug Discov. 5, 883–902 (2010).

    Article  CAS  PubMed  Google Scholar 

  113. Wu, Y. J. & Su, W. G. Recent developments on ketolides and macrolides. Curr. Med. Chem. 8, 1727–1758 (2001).

    Article  CAS  PubMed  Google Scholar 

  114. Nicolaou, K. C., Chen, J. S., Edmonds, D. J. & Estrada, A. A. Recent advances in the chemistry and biology of naturally occurring antibiotics. Angew. Chem. Int. Ed Engl. 48, 660–719 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Walsh, C. T., Acker, M. G. & Bowers, A. A. Thiazolyl peptide antibiotic biosynthesis: a cascade of post-translational modifications on ribosomal nascent proteins. J. Biol. Chem. 285, 27525–27531 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Brandi, L. et al. Specific, efficient, and selective inhibition of prokaryotic translation initiation by a novel peptide antibiotic. Proc. Natl Acad. Sci. USA 103, 39–44 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Llano-Sotelo, B., Klepacki, D. & Mankin, A. S. Selection of small peptides, inhibitors of translation. J. Mol. Biol. 391, 813–819 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Brandi, L. et al. Structural and functional characterization of the bacterial translocation inhibitor GE82832. FEBS Lett. 586, 3373–3378 (2012).

    Article  CAS  PubMed  Google Scholar 

  119. Fabbretti, A. et al. The antibiotic Furvina® targets the P-site of 30S ribosomal subunits and inhibits translation initiation displaying start codon bias. Nucleic Acids Res. 40, 10366–10374 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Maffioli, S. I. et al. Orthoformimycin, a selective inhibitor of bacterial translation elongation from Streptomyces containing an unusual orthoformate. ACS Chem. Biol. 8, 1939–1946 (2013).

    Article  CAS  PubMed  Google Scholar 

  121. Schroeder, S. J., Blaha, G., Tirado-Rives, J., Steitz, T. A. & Moore, P. B. The structures of antibiotics bound to the E site region of the 50 S ribosomal subunit of Haloarcula marismortui: 13-deoxytedanolide and girodazole. J. Mol. Biol. 367, 1471–1479 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Schneider-Poetsch, T. et al. Inhibition of eukaryotic translation elongation by cycloheximide and lactimidomycin. Nature Chem. Biol. 6, 209–217 (2010).

    Article  CAS  Google Scholar 

  123. Klinge, S., Voigts-Hoffmann, F., Leibundgut, M., Arpagaus, S. & Ban, N. Crystal structure of the eukaryotic 60S ribosomal subunit in complex with initiation factor 6. Science 334, 941–948 (2011). The structures of eukaryotic ribosomes reported in this work and in references 124–126 open the way to investigating the specificity of antibiotic interactions with the ribosome, as well as the development of anticancer drugs.

    Article  CAS  PubMed  Google Scholar 

  124. Ben-Shem, A. et al. The structure of the eukaryotic ribosome at 3.0 Å resolution. Science 334, 1524–1529 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Rabl, J., Leibundgut, M., Ataide, S. F., Haag, A. & Ban, N. Crystal structure of the eukaryotic 40S ribosomal subunit in complex with initiation factor 1. Science 331, 730–736 (2011).

    Article  CAS  PubMed  Google Scholar 

  126. Anger, A. M. et al. Structures of the human and Drosophila 80S ribosome. Nature 497, 80–85 (2013).

    Article  CAS  PubMed  Google Scholar 

  127. Keeling, K. M., Wang, D., Conard, S. E. & Bedwell, D. M. Suppression of premature termination codons as a therapeutic approach. Crit. Rev. Biochem. Mol. Biol. 47, 444–463 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Noller, H. F., Yusupov, M. M., Yusupova, G. Z., Baucom, A. & Cate, J. H. D. Translocation of tRNA during protein synthesis. FEBS Letter 514, 11–16 (2002).

    Article  CAS  Google Scholar 

  129. Rodnina, M. V. & Wintermeyer, W. The ribosome as a molecular machine: the mechanism of tRNA-mRNA movement in translocation. Biochem. Soc. Trans. 39, 658–662 (2011).

    Article  CAS  PubMed  Google Scholar 

  130. Chen, J., Tsai, A., O'Leary, S. E., Petrov, A. & Puglisi, J. D. Unraveling the dynamics of ribosome translocation. Curr. Opin. Struct. Biol. 22, 804–814 (2013).

    Article  CAS  Google Scholar 

  131. Moazed, D. & Noller, H. F. Intermediate states in the movement of transfer RNA in the ribosome. Nature 342, 142–148 (1989).

    Article  CAS  PubMed  Google Scholar 

  132. Munro, J. B., Altman, R. B., O'Connor, N. & Blanchard, S. C. Identification of two distinct hybrid state intermediates on the ribosome. Mol. Cell 25, 505–517 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Agirrezabala, X. et al. Visualization of the hybrid state of tRNA binding promoted by spontaneous ratcheting of the ribosome. Mol. Cell 32, 190–197 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Julian, P. et al. Structure of ratcheted ribosomes with tRNAs in hybrid states. Proc. Natl Acad. Sci. USA 105, 16924–16927 (2008).

    Article  PubMed  Google Scholar 

  135. Frank, J. & Agrawal, R. Ratchet-like movements between the two ribosomal subunits: their implications in elongation factor recognition and tRNA translocation. Cold Spring Harb. Symp. Quant. Biol. 66, 67–75 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  137. Savelsbergh, A. et al. An elongation factor G-induced ribosome rearrangement precedes tRNA-mRNA translocation. Mol. Cell 11, 1517–1523 (2003).

    Article  CAS  PubMed  Google Scholar 

  138. Ratje, A. H. et al. Head swivel on the ribosome facilitates translocation by means of intra-subunit tRNA hybrid sites. Nature 468, 713–716 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Borovinskaya, M. A., Shoji, S., Holton, J. M., Fredrick, K. & Cate, J. H. A steric block in translation caused by the antibiotic spectinomycin. ACS Chem. Biol. 2, 545–552 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Schluenzen, F. et al. The antibiotic kasugamycin mimics mRNA nucleotides to destabilize tRNA binding and inhibit canonical translation initiation. Nature Struct. Mol. Biol. 13, 871–878 (2006).

    Article  CAS  Google Scholar 

  141. Borovinskaya, M. A., Shoji, S., Fredrick, K. & Cate, J. H. Structural basis for hygromycin B inhibition of protein biosynthesis. RNA 14, 1590–1599 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Carter, A. P. et al. Functional insights from the structure of the 30S ribosomal subunit and its interactions with antibiotics. Nature 407, 340–348 (2000).

    Article  CAS  PubMed  Google Scholar 

  143. Hansen, J. L., Schmeing, T. M., Moore, P. B. & Steitz, T. A. Structural insights into peptide bond formation. Proc. Natl Acad. Sci. USA 99, 11670–11675 (2002).

    Article  CAS  PubMed  Google Scholar 

  144. Schlunzen, F., Pyetan, E., Fucini, P., Yonath, A. & Harms, J. Inhibition of peptide bond formation by pleuromutilins: the structure of the 50S ribosomal subunit from Deinococcus radiodurans in complex with tiamulin. Mol. Microbiol. 54, 1287–1294 (2004).

    Article  CAS  PubMed  Google Scholar 

  145. Seidelt, B. et al. Structural insight into nascent polypeptide chain-mediated translational stalling. Science 326, 1412–1415 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

I give special thanks to A. Mankin, B. Beatrix and members of my laboratory for critical reading of the manuscript. I also thank the Deutsche Forschungsgemeinschaft (WI3285/2-1) and the EMBO young investigator grant for funding research related to this topic.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel N. Wilson.

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

PowerPoint slides

Glossary

Peptidyl-transferase centre

(PTC). The site of peptide-bond formation on the large subunit, comprising universally conserved residues of domain V of the 23S ribosomal RNA.

Classical tRNA state

The tRNA binding state in which the anticodon and acceptor arms of the tRNA occupy the same sites on the 30S and 50S subunit, respectively.

Stacking interaction

Non-covalent interaction established by parallel plate stacked orientation of two moieties, with at least one being aromatic.

Non-cognate tRNAs

tRNAs that have a mismatched base-pairing interaction with mRNA codons. Accommodation leads to misincorporation of an incorrect amino acid into the nascent polypeptide chain.

Decoding centre

The A-site region of the small ribosomal subunit that is responsible for discriminating cognate from non-cognate tRNAs based on anticodon–codon interactions. This site encompasses the universally conserved nucleotides A1492 and A1493 of helix 44 of the 16S rRNA.

Cognate tRNAs

tRNAs that have a matched base-pairing interaction with mRNA codons. Accommodation leads to incorporation of the correct amino acid into the nascent polypeptide chain.

Intersubunit bridges

Sites of interaction at the interface between the small and large ribosomal subunits.

Hybrid state

The tRNA binding state in which the anticodon and acceptor arms of the tRNA occupy different sites on the 30S and 50S subunit, respectively.

Peptidyl-tRNA drop-off

Dissociation of tRNAs bearing the nascent polypeptide chain from the ribosome.

Bacteriocidal

Bacteriocidal antibiotics induce cell death, as exemplified by the aminoglycoside class of ribosome-targeting antibiotics.

Bacteriostatic

Bacteriostatic antibiotics inhibit bacterial growth but do not induce cell death, such that removal of the antibiotic leads to restoration of growth. Most ribosome-targeting antibiotics are bacteriostatic.

MATE family of transporters

(Multidrug and toxic compound extrusion family of transporters). MATE transporters couple antibiotic efflux (against their prevailing concentration gradient) to the energetically favourable movement of sodium ions (Na+) into the cell (along their electrochemical gradient).

Porins

β-barrel proteins that span the cell membrane and act as pores through which molecules can diffuse.

Compensatory mutations

In the context of this Review, secondary mutations that arise to restore bacterial fitness following a decrease in fitness owing to antibiotic resistance mutations.

Translation profiles

An indication of the changes in the levels of synthesis of proteins by ribosomes.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wilson, D. Ribosome-targeting antibiotics and mechanisms of bacterial resistance. Nat Rev Microbiol 12, 35–48 (2014). https://doi.org/10.1038/nrmicro3155

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro3155

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