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:

Molecular mechanisms of antibiotic resistance revisited

An Author Correction to this article was published on 02 February 2024

This article has been updated

Abstract

Antibiotic resistance is a global health emergency, with resistance detected to all antibiotics currently in clinical use and only a few novel drugs in the pipeline. Understanding the molecular mechanisms that bacteria use to resist the action of antimicrobials is critical to recognize global patterns of resistance and to improve the use of current drugs, as well as for the design of new drugs less susceptible to resistance development and novel strategies to combat resistance. In this Review, we explore recent advances in understanding how resistance genes contribute to the biology of the host, new structural details of relevant molecular events underpinning resistance, the identification of new resistance gene families and the interactions between different resistance mechanisms. Finally, we discuss how we can use this information to develop the next generation of antimicrobial therapies.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Overview of the molecular mechanisms of antibiotic resistance.
Fig. 2: Structure and entry channels of RND efflux systems.
Fig. 3: Antibiotic resistance via target protection, drug inactivation and target bypass.

Similar content being viewed by others

Change history

References

  1. Blair, J. M. A., Webber, M. A., Baylay, A. J., Ogbolu, D. O. & Piddock, L. J. V. Molecular mechanisms of antibiotic resistance. Nat. Rev. Microbiol. 13, 42–51 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Whittle, E. E. et al. Efflux impacts intracellular accumulation only in actively growing bacterial cells. mBio 12, e0260821 (2021).

    Article  PubMed  Google Scholar 

  3. Alav, I. et al. Structure, assembly, and function of tripartite efflux and type 1 secretion systems in gram-negative bacteria. Chem. Rev. 121, 5479–5596 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Klenotic, P. A., Morgan, C. E. & Yu, E. W. Cryo-EM as a tool to study bacterial efflux systems and the membrane proteome. Fac. Rev. 10, 24 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Malaka De Silva, P. et al. A tale of two plasmids: contributions of plasmid associated phenotypes to epidemiological success among Shigella. Proc. Biol. Sci. 289, 20220581 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Newbury, A. et al. Fitness effects of plasmids shape the structure of bacteria-plasmid interaction networks. Proc. Natl Acad. Sci. USA 119, e2118361119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Carrilero, L. et al. Positive selection inhibits plasmid coexistence in bacterial genomes. mBio 12, e00558-21 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Cummins, E. A., Snaith, A. E., McNally, A. & Hall, R. J. The role of potentiating mutations in the evolution of pandemic Escherichia coli clones. Eur. J. Clin. Microbiol. Infect. Dis. https://doi.org/10.1007/S10096-021-04359-3 (2021).

    Article  PubMed  Google Scholar 

  9. Gomez-Simmonds, A. & Uhlemann, A. C. Clinical implications of genomic adaptation and evolution of carbapenem-resistant Klebsiella pneumoniae. J. Infect. Dis. 215, S18–S27 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mishra, N. N. et al. Daptomycin resistance in enterococci is associated with distinct alterations of cell membrane phospholipid content. PLoS ONE 7, e43958 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Draper, P. The outer parts of the mycobacterial envelope as permeability barriers. Front. Biosci. 3, D1253-61 (1998).

    Article  PubMed  Google Scholar 

  12. Nikaido, H. Molecular basis of bacterial outer membrane permeability revisited. Microbiol. Mol. Biol. Rev. 67, 593–656 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fernández, L. & Hancock, R. E. W. Adaptive and mutational resistance: role of porins and efflux pumps in drug resistance. Clin. Microbiol. Rev. 25, 661–681 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Baslé, A., Rummel, G., Storici, P., Rosenbusch, J. P. & Schirmer, T. Crystal structure of osmoporin OmpC from E. coli at 2.0 Å. J. Mol. Biol. 362, 933–942 (2006).

    Article  PubMed  Google Scholar 

  15. Acosta-Gutiérrez, S. et al. Getting drugs into gram-negative bacteria: rational rules for permeation through general porins. ACS Infect. Dis. 4, 1487–1498 (2018).

    Article  PubMed  Google Scholar 

  16. Wong, J. L. C. et al. OmpK36-mediated Carbapenem resistance attenuates ST258 Klebsiella pneumoniae in vivo. Nat. Commun. 10, 3957 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  17. Lou, H. et al. Altered antibiotic transport in OmpC mutants isolated from a series of clinical strains of multi-drug resistant E. coli. PLoS ONE 6, e25825 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pratt, L. A., Hsing, W., Gibson, K. E. & Silhavy, T. J. From acids to osmZ: multiple factors influence synthesis of the OmpF and OmpC porins in Escherichia coli. Mol. Microbiol. 20, 911–917 (1996).

    Article  CAS  PubMed  Google Scholar 

  19. Adler, M., Anjum, M., Andersson, D. I. & Sandegren, L. Influence of acquired β-lactamases on the evolution of spontaneous carbapenem resistance in Escherichia coli. J. Antimicrob. Chemother. 68, 51–59 (2013).

    Article  CAS  PubMed  Google Scholar 

  20. Andersen, J. & Delihas, N. micF RNA binds to the 5’ end of ompF mRNA and to a protein from Escherichia coli. Biochemistry 29, 9249–9256 (1990).

    Article  CAS  PubMed  Google Scholar 

  21. Delihas, N. & Forst, S. MicF: an antisense RNA gene involved in response of Escherichia coli to global stress factors. J. Mol. Biol. 313, 1–12 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Chen, S., Zhang, A., Blyn, L. B. & Storz, G. MicC, a second small-RNA regulator of Omp protein expression in Escherichia coli. J. Bacteriol. 186, 6689–6697 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dam, S., Pagès, J.-M. & Masi, M. Dual regulation of the small RNA MicC and the quiescent porin OmpN in response to antibiotic stress in Escherichia coli. Antibiotics 6, 33 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Eren, E. et al. Substrate specificity within a family of outer membrane carboxylate channels. PLoS Biol. 10, e1001242 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zgurskaya, H. I. & Rybenkov, V. V. Permeability barriers of Gram-negative pathogens. Ann. NY Acad. Sci. 1459, 5–18 (2020).

    Article  ADS  PubMed  Google Scholar 

  26. Chevalier, S. et al. Structure, function and regulation of Pseudomonas aeruginosa porins. FEMS Microbiol. Rev. 41, 698–722 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Ude, J. et al. Outer membrane permeability: antimicrobials and diverse nutrients bypass porins in Pseudomonas aeruginosa. Proc. Natl Acad. Sci. USA 118, e2107644118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nazarov, P. A. MDR pumps as crossroads of resistance: antibiotics and bacteriophages. Antibiotics 11, 734 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tsutsumi, K. et al. Structures of the wild-type MexAB–OprM tripartite pump reveal its complex formation and drug efflux mechanism. Nat. Commun. 10, 1520 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  30. Du, D. et al. Multidrug efflux pumps: structure, function and regulation. Nat. Rev. Microbiol. 16, 523–539 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Ebbensgaard, A. E., Løbner-Olesen, A. & Frimodt-Møller, J. The role of efflux pumps in the transition from low-level to clinical antibiotic resistance. Antibiotics 9, 855 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Morgan, C. E. et al. Cryoelectron microscopy structures of AdeB illuminate mechanisms of simultaneous binding and exporting of substrates. mBio 12, e03690-20 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Chen, M. et al. In situ structure of the AcrAB-TolC efflux pump at subnanometer resolution. Structure https://doi.org/10.1016/J.STR.2021.08.008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Wang, Z. et al. An allosteric transport mechanism for the AcrAB-TolC multidrug efflux pump. eLife 6, e24905 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Tikhonova, E. B., Yamada, Y. & Zgurskaya, H. I. Sequential mechanism of assembly of multidrug efflux pump AcrAB-TolC. Chem. Biol. 18, 454–463 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. López, C. A., Travers, T., Pos, K. M., Zgurskaya, H. I. & Gnanakaran, S. Dynamics of intact MexAB-OprM efflux pump: focusing on the MexA-OprM interface. Sci. Rep. 7, 16521 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  37. Du, D. et al. Structure of the AcrAB–TolC multidrug efflux pump. Nature 509, 512–515 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jo, I. et al. Recent paradigm shift in the assembly of bacterial tripartite efflux pumps and the type I secretion system. J. Microbiol. 57, 185–194 (2019).

    Article  PubMed  Google Scholar 

  39. Glavier, M. et al. Antibiotic export by MexB multidrug efflux transporter is allosterically controlled by a MexA-OprM chaperone-like complex. Nat. Commun. 11, 4948 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bavro, V. N., Marshall, R. L. & Symmons, M. F. Architecture and roles of periplasmic adaptor proteins in tripartite efflux assemblies. Front. Microbiol. 6, 513 (2015).

    PubMed  PubMed Central  Google Scholar 

  41. McNeil, H. E. et al. Identification of binding residues between periplasmic adapter protein (PAP) and RND efflux pumps explains PAP-pump promiscuity and roles in antimicrobial resistance. PLoS Pathog. 15, e1008101 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Abdali, N. et al. Reviving antibiotics: efflux pump inhibitors that interact with AcrA, a membrane fusion protein of the AcrAB-TolC multidrug efflux pump. ACS Infect. Dis. 3, 89–98 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Salehi, B., Ghalavand, Z., Yadegar, A. & Eslami, G. Characteristics and diversity of mutations in regulatory genes of resistance-nodulation-cell division efflux pumps in association with drug-resistant clinical isolates of Acinetobacter baumannii. Antimicrob. Resist. Infect. Control 10, 53 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Shafer, W. M. et al. in Efflux-Mediated Antimicrobial Resistance in Bacteria (eds Li, X.-Z., Elkins, C. A. & Zgurskaya, H. I.) 439–469 (Adis, 2016).

  45. Kobylka, J., Kuth, M. S., Müller, R. T., Geertsma, E. R. & Pos, K. M. AcrB: a mean, keen, drug efflux machine. Ann. NY Acad. Sci. 1459, 38–68 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  46. Zwama, M. & Nishino, K. Ever-adapting RND efflux pumps in Gram-negative multidrug-resistant pathogens: a race against time. Antibiotics 10, 774 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zwama, M. et al. Multiple entry pathways within the efflux transporter AcrB contribute to multidrug recognition. Nat. Commun. 9, 124 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  48. Tam, H.-K. et al. Allosteric drug transport mechanism of multidrug transporter AcrB. Nat. Commun. 12, 3889 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Nakashima, R., Sakurai, K., Yamasaki, S., Nishino, K. & Yamaguchi, A. Structures of the multidrug exporter AcrB reveal a proximal multisite drug-binding pocket. Nature 480, 565–569 (2011).

    Article  ADS  CAS  PubMed  Google Scholar 

  50. Tam, H.-K. et al. Binding and transport of carboxylated drugs by the multidrug transporter AcrB. J. Mol. Biol. 432, 861 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Hobbs, E. C., Yin, X., Paul, B. J., Astarita, J. L. & Storz, G. Conserved small protein associates with the multidrug efflux pump AcrB and differentially affects antibiotic resistance. Proc. Natl Acad. Sci. USA 109, 16696–16701 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Du, D. et al. Interactions of a bacterial RND transporter with a transmembrane small protein in a lipid environment. Structure 28, 625 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Venter, H., Mowla, R., Ohene-Agyei, T. & Ma, S. RND-type drug efflux pumps from Gram-negative bacteria: molecular mechanism and inhibition. Front. Microbiol. 06, 377 (2015).

    Article  Google Scholar 

  54. Aron, Z. & Opperman, T. J. The hydrophobic trap — the Achilles heel of RND efflux pumps. Res. Microbiol. 169, 393–400 (2018).

    Article  PubMed  Google Scholar 

  55. Gerson, S. et al. Diversity of mutations in regulatory genes of resistance-nodulation-cell division efflux pumps in association with tigecycline resistance in Acinetobacter baumannii. J. Antimicrob. Chemother. 73, 1501–1508 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. Veal, W. L., Nicholas, R. A. & Shafer, W. M. Overexpression of the MtrC-MtrD-MtrE efflux pump due to an mtrR mutation is required for chromosomally mediated penicillin resistance in Neisseria gonorrhoeae. J. Bacteriol. 184, 5619–5624 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen, S. et al. Could dampening expression of the Neisseria gonorrhoeae mtrCDE-encoded efflux pump be a strategy to preserve currently or resurrect formerly used antibiotics to treat gonorrhea? mBio 10, e01576-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zarantonelli, L., Borthagaray, G., Lee, E.-H. & Shafer, W. M. Decreased azithromycin susceptibility of Neisseria gonorrhoeae due to mtrR mutations. Antimicrob. Agents Chemother. 43, 2468–2472 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Handing, J. W., Ragland, S. A., Bharathan, U. V. & Criss, A. K. The MtrCDE efflux pump contributes to survival of Neisseria gonorrhoeae from human neutrophils and their antimicrobial components. Front. Microbiol. 9, 2688 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Wadsworth, C. B., Arnold, B. J., Sater, M. R. A. & Grad, Y. H. Azithromycin resistance through interspecific acquisition of an epistasis-dependent efflux pump component and transcriptional regulator in Neisseria gonorrhoeae. mBio 9, e01419-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Castanheira, M., Doyle, T. B., Smith, C. J., Mendes, R. E. & Sader, H. S. Combination of MexAB-OprM overexpression and mutations in efflux regulators, PBPs and chaperone proteins is responsible for ceftazidime/avibactam resistance in Pseudomonas aeruginosa clinical isolates from US hospitals. J. Antimicrob. Chemother. 74, 2588–2595 (2019).

    Article  CAS  PubMed  Google Scholar 

  62. Grinnage-Pulley, T. & Zhang, Q. Genetic basis and functional consequences of differential expression of the CmeABC efflux pump in Campylobacter jejuni isolates. PLoS ONE 10, e0131534 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Grimsey, E. M., Weston, N., Ricci, V., Stone, J. W. & Piddock, L. J. V. Overexpression of RamA, which regulates production of the multidrug resistance efflux pump AcrAB-TolC, increases mutation rate and influences drug resistance phenotype. Antimicrob. Agents Chemother. 64, e02460-19 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Yamasaki, S. et al. Crystal structure of the multidrug resistance regulator RamR complexed with bile acids. Sci. Rep. 9, 177 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  65. Duval, V. & Lister, I. M. MarA, SoxS and Rob of Escherichia coli – global regulators of multidrug resistance, virulence and stress response. Int. J. Biotechnol. Wellness Ind. 2, 101 (2013).

    PubMed  PubMed Central  Google Scholar 

  66. Alav, I., Sutton, J. M. & Rahman, K. M. Role of bacterial efflux pumps in biofilm formation. J. Antimicrob. Chemother. 73, 2003–2020 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Holden, E. & Webber, M. Defining the link between efflux pumps and biofilm formation. Access Microbiol. https://doi.org/10.1099/acmi.mim2019.po0007 (2020).

  68. Sharma, P. et al. The multiple antibiotic resistance operon of enteric bacteria controls DNA repair and outer membrane integrity. Nat. Commun. 8, 1444 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  69. Housseini B Issa, K., Phan, G. & Broutin, I. Functional mechanism of the efflux pumps transcription regulators from Pseudomonas aeruginosa based on 3D structures. Front. Mol. Biosci. 5, 57 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Sharma, A., Gupta, V. K. & Pathania, R. Efflux pump inhibitors for bacterial pathogens: from bench to bedside. Indian J. Med. Res. 149, 129 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Wang, Y., Venter, H. & Ma, S. Efflux pump inhibitors: a novel approach to combat efflux-mediated drug resistance in bacteria. Curr. Drug Targets 17, 702–719 (2016).

    Article  CAS  PubMed  Google Scholar 

  72. Pule, C. M. et al. Efflux pump inhibitors: targeting mycobacterial efflux systems to enhance TB therapy. J. Antimicrob. Chemother. 71, 17–26 (2016).

    Article  CAS  PubMed  Google Scholar 

  73. Machado, D. et al. Interplay between mutations and efflux in drug resistant clinical isolates of Mycobacterium tuberculosis. Front. Microbiol. 8, 711 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Zimmermann, S. et al. Clinically approved drugs inhibit the Staphylococcus aureus multidrug NorA efflux pump and reduce biofilm formation. Front. Microbiol. 10, 2762 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Baquero, F. & Levin, B. R. Proximate and ultimate causes of the bactericidal action of antibiotics. Nat. Rev. Microbiol. 19, 123–132 (2021).

    Article  CAS  PubMed  Google Scholar 

  76. Bush, N. G., Diez-Santos, I., Abbott, L. R. & Maxwell, A. Quinolones: mechanism, lethality and their contributions to antibiotic resistance. Molecules 25, 5662 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Periasamy, H. et al. High prevalence of Escherichia coli clinical isolates in India harbouring four amino acid inserts in PBP3 adversely impacting activity of aztreonam/avibactam. J. Antimicrob. Chemother. 75, 1650–1651 (2020).

    Article  CAS  PubMed  Google Scholar 

  78. Huber, S. et al. Genomic and phenotypic analysis of linezolid-resistant Staphylococcus epidermidis in a Tertiary Hospital in Innsbruck, Austria. Microorganisms 9, 1023 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Alfsnes, K. et al. A genomic view of experimental intraspecies and interspecies transformation of a rifampicin-resistance allele into Neisseria meningitidis. Microb. Genomics 4, e000222 (2018).

    Article  Google Scholar 

  80. Panda, A., Drancourt, M., Tuller, T. & Pontarotti, P. Genome-wide analysis of horizontally acquired genes in the genus Mycobacterium. Sci. Rep. 8, 14817 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  81. Bhagwat, A., Deshpande, A. & Parish, T. How Mycobacterium tuberculosis drug resistance has shaped anti-tubercular drug discovery. Front. Cell. Infect. Microbiol. 12, 974101 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sun, Q. et al. The molecular basis of pyrazinamide activity on Mycobacterium tuberculosis PanD. Nat. Commun. 11, 339 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bhujbalrao, R. & Anand, R. Deciphering determinants in ribosomal methyltransferases that confer antimicrobial resistance. J. Am. Chem. Soc. 141, 1425–1429 (2019).

    Article  CAS  PubMed  Google Scholar 

  84. Doi, Y., Wachino, J. I. & Arakawa, Y. Aminoglycoside resistance: the emergence of acquired 16S ribosomal RNA methyltransferases. Infect. Dis. Clin. 30, 523–537 (2016).

    Article  Google Scholar 

  85. Elias, R., Duarte, A. & Perdigão, J. A molecular perspective on colistin and Klebsiella pneumoniae: mode of action, resistance genetics, and phenotypic susceptibility. Diagnostics 11, 1165 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Sabnis, A. et al. Colistin kills bacteria by targeting lipopolysaccharide in the cytoplasmic membrane. eLife 10, e65836 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Huang, J. et al. Regulating polymyxin resistance in Gram-negative bacteria: roles of two-component systems PhoPQ and PmrAB. Future Microbiol. 15, 445–459 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hamel, M., Rolain, J.-M. & Baron, S. A. The history of colistin resistance mechanisms in bacteria: progress and challenges. Microorganisms 9, 442 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Xu, Y. et al. An evolutionarily conserved mechanism for intrinsic and transferable polymyxin resistance. mBio 9, e02317-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Liao, W. et al. High prevalence of colistin resistance and mcr-9/10 genes in Enterobacter spp. in a tertiary hospital over a decade. Int. J. Antimicrob. Agents 59, 106573 (2022).

    Article  CAS  PubMed  Google Scholar 

  91. Zhu, X. Q. et al. Impact of mcr-1 on the development of high level colistin resistance in Klebsiella pneumoniae and Escherichia coli. Front. Microbiol. 12, 878 (2021).

    Google Scholar 

  92. Purcell, A. B., Voss, B. J. & Trent, M. S. Diacylglycerol kinase A is essential for polymyxin resistance provided by EptA, MCR-1, and other lipid A phosphoethanolamine transferases. J. Bacteriol. 204, e0049821 (2022).

    Article  PubMed  Google Scholar 

  93. Yang, Q. et al. Balancing mcr-1 expression and bacterial survival is a delicate equilibrium between essential cellular defence mechanisms. Nat. Commun. 8, 2054 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  94. Ruiz, J. Transferable mechanisms of quinolone resistance from 1998 onward. Clin. Microbiol. Rev. 32, e00007-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  95. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  96. Crowe-McAuliffe, C. et al. Structural basis of ABCF-mediated resistance to pleuromutilin, lincosamide, and streptogramin A antibiotics in Gram-positive pathogens. Nat. Commun. 12, 3577 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  97. Murina, V., Kasari, M., Hauryliuk, V. & Atkinson, G. C. Antibiotic resistance ABCF proteins reset the peptidyl transferase centre of the ribosome to counter translational arrest. Nucleic Acids Res. 46, 3753 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Su, W. et al. Ribosome protection by antibiotic resistance ATP-binding cassette protein. Proc. Natl Acad. Sci. USA 115, 5157–5162 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  99. Ero, R., Kumar, V., Su, W. & Gao, Y.-G. Ribosome protection by ABC-F proteins — molecular mechanism and potential drug design. Protein Sci. 28, 684–693 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Mohamad, M. et al. Sal-type ABC-F proteins: intrinsic and common mediators of pleuromutilin resistance by target protection in staphylococci. Nucleic Acids Res. 50, 2128–2142 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Forsberg, K. J., Patel, S., Wencewicz, T. A. & Dantas, G. The tetracycline destructases: a novel family of tetracycline-inactivating enzymes. Chem. Biol. 22, 888–897 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Schaenzer, A. J. & Wright, G. D. Antibiotic resistance by enzymatic modification of antibiotic targets. Trends Mol. Med. 26, 768–782 (2020).

    Article  CAS  PubMed  Google Scholar 

  103. Tooke, C. L. et al. β-Lactamases and β-lactamase inhibitors in the 21st century. J. Mol. Biol. 431, 3472–3500 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Naas, T. et al. Beta-lactamase database (BLDB) – structure and function. J. Enzym. Inhib. Med. Chem. 32, 917–919 (2017).

    Article  CAS  Google Scholar 

  105. Ambler, R. P. The structure of β-lactamases. Philos. Trans. R. Soc. Lond. B Biol. Sci. 289, 321–331 (1980).

    Article  ADS  CAS  PubMed  Google Scholar 

  106. Bush, K. & Jacoby, G. A. Updated functional classification of β-lactamases. Antimicrob. Agents Chemother. 54, 969–976 (2010).

    Article  CAS  PubMed  Google Scholar 

  107. Lima, L. M., Silva, B. N. M. D., Barbosa, G. & Barreiro, E. J. β-Lactam antibiotics: an overview from a medicinal chemistry perspective. Eur. J. Med. Chem. 208, 112829 (2020).

    Article  CAS  PubMed  Google Scholar 

  108. Nepal, K. et al. Extended spectrum beta-lactamase and metallo beta-lactamase production among Escherichia coli and Klebsiella pneumoniae isolated from different clinical samples in a tertiary care hospital in Kathmandu, Nepal. Ann. Clin. Microbiol. Antimicrob. 16, 62 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  109. World Health Organization. WHO Publishes List of Bacteria for which New Antibiotics are Urgently Needed. World Health Organization https://www.who.int/news/item/27-02-2017-who-publishes-list-of-bacteria-for-which-new-antibiotics-are-urgently-needed (2017).

  110. Queenan, A. M. & Bush, K. Carbapenemases: the versatile β-lactamases. Clin. Microbiol. Rev. 20, 440–458 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Yoon, E.-J. et al. A novel KPC variant KPC-55 in Klebsiella pneumoniae ST307 of reinforced meropenem-hydrolyzing activity. Front. Microbiol. 11, 2509 (2020).

    Article  Google Scholar 

  112. Mancini, S., Keller, P. M., Greiner, M., Bruderer, V. & Imkamp, F. Detection of NDM-19, a novel variant of the New Delhi metallo-β-lactamase with increased carbapenemase activity under zinc-limited conditions, in Switzerland. Diagn. Microbiol. Infect. Dis. 95, 114851 (2019).

    Article  CAS  PubMed  Google Scholar 

  113. Tietgen, M. et al. Identification of the novel class D β-lactamase OXA-679 involved in carbapenem resistance in Acinetobacter calcoaceticus. J. Antimicrob. Chemother. 74, 1494–1502 (2019).

    Article  CAS  PubMed  Google Scholar 

  114. Yong, D. et al. Characterization of a new metallo-β-lactamase Gene, blaNDM-1, and a novel erythromycin esterase gene carried on a unique genetic structure in Klebsiella pneumoniae sequence type 14 from India. Antimicrob. Agents Chemother. 53, 5046–5054 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Johnson, A. P. & Woodford, N. Global spread of antibiotic resistance: the example of New Delhi metallo-β-lactamase (NDM)-mediated carbapenem resistance. J. Med. Microbiol. 62, 499–513 (2013).

    Article  CAS  PubMed  Google Scholar 

  116. Li, X. et al. Dissemination of bla NDM-5 gene via an IncX3-type plasmid among non-clonal Escherichia coli in China. Antimicrob. Resist. Infect. Control 7, 59 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Pillonetto, M. et al. First report of NDM-1-producing Acinetobacter baumannii sequence type 25 in Brazil. Antimicrob. Agents Chemother. 58, 7592–7594 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Principe, L. et al. First report of NDM-1-producing Klebsiella pneumoniae imported from Africa to Italy: evidence of the need for continuous surveillance. J. Glob. Antimicrob. Resist. 8, 23–27 (2017).

    Article  PubMed  Google Scholar 

  119. D’Souza, A. W. et al. Spatiotemporal dynamics of multidrug resistant bacteria on intensive care unit surfaces. Nat. Commun. 10, 4569 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  120. Carattoli, A. Plasmids in Gram negatives: molecular typing of resistance plasmids. Int. J. Med. Microbiol. 301, 654–658 (2011).

    Article  CAS  PubMed  Google Scholar 

  121. Hammoudi Halat, D. & Ayoub Moubareck, C. The current burden of carbapenemases: review of significant properties and dissemination among gram-negative bacteria. Antibiotics 9, 186 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Bush, K. Alarming β-lactamase-mediated resistance in multidrug-resistant Enterobacteriaceae. Curr. Opin. Microbiol. 13, 558–564 (2010).

    Article  CAS  PubMed  Google Scholar 

  123. Chatterjee, S. et al. Carbapenem resistance in Acinetobacter baumannii and other Acinetobacter spp. causing neonatal sepsis: focus on NDM-1 and Its linkage to ISAba125. Front. Microbiol. 7, 1126 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  124. Héritier, C., Poirel, L. & Nordmann, P. Cephalosporinase over-expression resulting from insertion of ISAba1 in Acinetobacter baumannii. Clin. Microbiol. Infect. 12, 123–130 (2006).

    Article  PubMed  Google Scholar 

  125. Fang, L. et al. Emerging high‐level tigecycline resistance: novel tetracycline destructases spread via the mobile Tet(X). BioEssays 42, 2000014 (2020).

    Article  CAS  Google Scholar 

  126. Gasparrini, A. J. et al. Tetracycline-inactivating enzymes from environmental, human commensal, and pathogenic bacteria cause broad-spectrum tetracycline resistance. Commun. Biol. 3, 241 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. He, T. et al. Emergence of plasmid-mediated high-level tigecycline resistance genes in animals and humans. Nat. Microbiol. 4, 1450–1456 (2019).

    Article  CAS  PubMed  Google Scholar 

  128. Sun, J. et al. Plasmid-encoded tet(X) genes that confer high-level tigecycline resistance in Escherichia coli. Nat. Microbiol. 4, 1457–1464 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Wang, L. et al. Novel plasmid-mediated tet (X5) gene conferring resistance to tigecycline, eravacycline, and omadacycline in a clinical Acinetobacter baumannii isolate. Antimicrob. Agents Chemother. 64, e01326-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Szychowski, J. et al. Inhibition of aminoglycoside-deactivating enzymes APH(3’)-IIIa and AAC(6’)-Ii by amphiphilic paromomycin O2”-ether analogues. ChemMedChem 6, 1961–1966 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  132. Bordeleau, E. et al. ApmA is a unique aminoglycoside antibiotic acetyltransferase that inactivates apramycin. mBio 12, e02705-20 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Feßler, A. T., Wang, Y., Wu, C. & Schwarz, S. Mobile lincosamide resistance genes in staphylococci. Plasmid 99, 22–31 (2018).

    Article  PubMed  Google Scholar 

  134. Zhu, X.-Q. et al. Novel lnu(G) gene conferring resistance to lincomycin by nucleotidylation, located on Tn6260 from Enterococcus faecalis E531. J. Antimicrob. Chemother. 72, 993–997 (2016).

    Google Scholar 

  135. Golkar, T., Zieliński, M. & Berghuis, A. M. Look and outlook on enzyme-mediated macrolide resistance. Front. Microbiol. 9, 1942 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Gu Liu, C. et al. Phage-antibiotic synergy is driven by a unique combination of antibacterial mechanism of action and stoichiometry. mBio 11, e01462-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  138. Luthra, S., Rominski, A. & Sander, P. The role of antibiotic-target-modifying and antibiotic-modifying enzymes in Mycobacterium abscessus drug resistance. Front. Microbiol. 9, 2179 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Rominski, A., Roditscheff, A., Selchow, P., Böttger, E. C. & Sander, P. Intrinsic rifamycin resistance of Mycobacterium abscessus is mediated by ADP-ribosyltransferase MAB_0591. J. Antimicrob. Chemother. 72, 376–384 (2017).

    Article  CAS  PubMed  Google Scholar 

  140. Surette, M. D., Spanogiannopoulos, P. & Wright, G. D. The enzymes of the rifamycin antibiotic resistome. Acc. Chem. Res. 54, 2065–2075 (2021).

    Article  CAS  PubMed  Google Scholar 

  141. Spanogiannopoulos, P., Thaker, M., Koteva, K., Waglechner, N. & Wright, G. D. Characterization of a rifampin-inactivating glycosyltransferase from a screen of environmental actinomycetes. Antimicrob. Agents Chemother. 56, 5061–5069 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Spanogiannopoulos, P., Waglechner, N., Koteva, K. & Wright, G. D. A rifamycin inactivating phosphotransferase family shared by environmental and pathogenic bacteria. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.1402358111 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  143. Stogios, P. J. et al. Rifampin phosphotransferase is an unusual antibiotic resistance kinase. Nat. Commun. 7, 11343 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  144. Koteva, K. et al. Rox, a rifamycin resistance enzyme with an unprecedented mechanism of action. Cell Chem. Biol. 25, 403–412.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  145. Munita, J. M. & Arias, C. A. Mechanisms of antibiotic resistance. Microbiol. Spectr. 23, 464–472 (2016).

    Google Scholar 

  146. Stapleton, P. D. & Taylor, P. W. Methicillin resistance in Staphylococcus aureus: mechanisms and modulation. Sci. Prog. 85, 57 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Larsen, J. et al. Emergence of methicillin resistance predates the clinical use of antibiotics. Nature 602, 135–141 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  148. Caveney, N. A. et al. Structural insight into YcbB-mediated beta-lactam resistance in Escherichia coli. Nat. Commun. 10, 1849 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  149. Hugonnet, J. E. et al. Factors essential for L,D-transpeptidase-mediated peptidoglycan cross-linking and β-lactam resistance in Escherichia coli. eLife 5, 19469 (2016).

    Article  Google Scholar 

  150. Gardete, S. & Tomasz, A. Mechanisms of vancomycin resistance in Staphylococcus aureus. J. Clin. Invest. 124, 2836–2840 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Arthur, M., Reynolds, P. & Courvalin, P. Glycopeptide resistance in enterococci. Trends Microbiol. 4, 401–407 (1996).

    Article  CAS  PubMed  Google Scholar 

  152. Miller, W. R., Munita, J. M. & Arias, C. A. Mechanisms of antibiotic resistance in enterococci. Expert Rev. Anti. Infect. Ther. 12, 1221–1236 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Sievert, D. M. et al. Vancomycin-Resistant Staphylococcus aureus in the United States, 2002–2006. Clin. Infect. Dis. 46, 668–674 (2008).

    Article  CAS  PubMed  Google Scholar 

  154. Melo-Cristino, J., Resina, C., Manuel, V., Lito, L. & Ramirez, M. First case of infection with vancomycin-resistant Staphylococcus aureus in Europe. Lancet 382, 205 (2013).

    Article  PubMed  Google Scholar 

  155. Martin, J. N. et al. Emergence of Trimethoprim-Sulfamethoxazole resistance in the AIDS era. J. Infect. Dis. 180, 1809–1818 (1999).

    Article  CAS  PubMed  Google Scholar 

  156. Bermingham, A. & Derrick, J. P. The folic acid biosynthesis pathway in bacteria: evaluation of potential for antibacterial drug discovery. BioEssays 24, 637–648 (2002).

    Article  CAS  PubMed  Google Scholar 

  157. Eliopoulos, G. M. & Huovinen, P. Resistance to trimethoprim-sulfamethoxazole. Clin. Infect. Dis. 32, 1608–1614 (2001).

    Article  Google Scholar 

  158. Jaeger, T. & Mayer, C. N-acetylmuramic acid 6-phosphate lyases (MurNAc etherases): role in cell wall metabolism, distribution, structure, and mechanism. Cell. Mol. Life Sci. 65, 928–939 (2008).

    Article  CAS  PubMed  Google Scholar 

  159. Gisin, J., Schneider, A., Nägele, B., Borisova, M. & Mayer, C. A cell wall recycling shortcut that bypasses peptidoglycan de novo biosynthesis. Nat. Chem. Biol. 9, 491–493 (2013).

    Article  CAS  PubMed  Google Scholar 

  160. Mayer, C. et al. Bacteria’s different ways to recycle their own cell wall. Int. J. Med. Microbiol. 309, 151326 (2019).

    Article  CAS  PubMed  Google Scholar 

  161. Meyer, B. & Cookson, B. Does microbial resistance or adaptation to biocides create a hazard in infection prevention and control? J. Hosp. Infect. 76, 200–205 (2010).

    Article  CAS  PubMed  Google Scholar 

  162. Papp-Wallace, K. M., Docquier, J. D., Kerff, F. & Power, P. Editorial: structural and biochemical aspects of the interaction of β-lactamases with state-of-the-art inhibitors. Front. Microbiol. 13, 849324 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  163. Boehr, D. D. et al. Broad-spectrum peptide inhibitors of aminoglycoside antibiotic resistance enzymes. Chem. Biol. 10, 189–196 (2003).

    Article  CAS  PubMed  Google Scholar 

  164. Lin, L. et al. Azithromycin synergizes with cationic antimicrobial peptides to exert bactericidal and therapeutic activity against highly multidrug-resistant Gram-negative bacterial pathogens. eBioMedicine 2, 690–698 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  165. Higgins, M. K. et al. Structure of the ligand-blocked periplasmic entrance of the bacterial multidrug efflux protein TolC. J. Mol. Biol. 342, 697–702 (2004).

    Article  CAS  PubMed  Google Scholar 

  166. Darzynkiewicz, Z. M. et al. Identification of binding sites for efflux pump inhibitors of the AcrAB-TolC component AcrA. Biophys. J. 116, 648–658 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  167. Ayhan, D. H. et al. Sequence-specific targeting of bacterial resistance genes increases antibiotic efficacy. PLoS Biol. 14, e1002552 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  168. Xu, Z. et al. Native CRISPR-Cas-mediated genome editing enables dissecting and sensitizing clinical multidrug-resistant P. aeruginosa. Cell Rep. 29, 1707–1717.e3 (2019).

    Article  CAS  PubMed  Google Scholar 

  169. Davis, B. D., Chen, L. L. & Tai, P. C. Misread protein creates membrane channels: an essential step in the bactericidal action of aminoglycosides. Proc. Natl Acad. Sci. USA 83, 6164–6168 (1986).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  170. Wachino, J.-I., Doi, Y. & Arakawa, Y. Aminoglycoside resistance: updates with a focus on acquired 16S ribosomal RNA methyltransferases. Infect. Dis. Clin. North Am. 34, 887–902 (2020).

    Article  PubMed  Google Scholar 

  171. Doi, Y., Wachino, J.-I. & Arakawa, Y. Aminoglycoside resistance: the emergence of acquired 16S ribosomal RNA methyltransferases. Infect. Dis. Clin. North Am. 30, 523–537 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  172. Pachori, P., Gothalwal, R. & Gandhi, P. Emergence of antibiotic resistance Pseudomonas aeruginosa in intensive care unit; a critical review. Genes Dis. 6, 109–119 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  173. Ur Rahman, S. et al. The growing genetic and functional diversity of extended spectrum beta-lactamases. Biomed. Res. Int. 2018, 1–14 (2018).

    Article  Google Scholar 

  174. Zapun, A., Contreras-Martel, C. & Vernet, T. Penicillin-binding proteins and β-lactam resistance. FEMS Microbiol. Rev. 32, 361–385 (2008).

    Article  CAS  PubMed  Google Scholar 

  175. Andrade, F. F., Silva, D., Rodrigues, A. & Pina-Vaz, C. Colistin update on its mechanism of action and resistance, present and future challenges. Microorganisms 8, 1716 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Liu, Y.-Y. et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: a microbiological and molecular biological study. Lancet Infect. Dis. 16, 161–168 (2016).

    Article  PubMed  Google Scholar 

  177. Moffatt, J. H. et al. Colistin resistance in Acinetobacter baumannii is mediated by complete loss of lipopolysaccharide production. Antimicrob. Agents Chemother. 54, 4971–4977 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Zeng, D. et al. Approved glycopeptide antibacterial drugs: mechanism of action and resistance. Cold Spring Harb. Perspect. Med. 6, a026989 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  179. Stogios, P. J. & Savchenko, A. Molecular mechanisms of vancomycin resistance. Protein Sci. 29, 654–669 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Hiramatsu, K. et al. Methicillin-resistant Staphylococcus aureus clinical strain with reduced vancomycin susceptibility. J. Antimicrob. Chemother. 40, 135–136 (1997).

    Article  CAS  PubMed  Google Scholar 

  181. Spížek, J. & Řezanka, T. Lincosamides: chemical structure, biosynthesis, mechanism of action, resistance, and applications. Biochem. Pharmacol. 133, 20–28 (2017).

    Article  PubMed  Google Scholar 

  182. 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 

  183. Novotna, G. & Janata, J. A new evolutionary variant of the streptogramin A resistance protein, Vga(A) LC, from Staphylococcus haemolyticus with shifted substrate specificity towards lincosamides. Antimicrob. Agents Chemother. 50, 4070–4076 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Jerala, R. Synthetic lipopeptides: a novel class of anti-infectives. Expert Opin. Investig. Drugs 16, 1159–1169 (2007).

    Article  CAS  PubMed  Google Scholar 

  185. Tran, T. T., Munita, J. M. & Arias, C. A. Mechanisms of drug resistance: daptomycin resistance. Ann. NY Acad. Sci. 1354, 32–53 (2015).

    Article  ADS  PubMed  Google Scholar 

  186. Vázquez-Laslop, N. & Mankin, A. S. How macrolide antibiotics work. Trends Biochem. Sci. 43, 668–684 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  188. Roberts, M. C. Update on macrolide-lincosamide-streptogramin, ketolide, and oxazolidinone resistance genes. FEMS Microbiol. Lett. 282, 147–159 (2008).

    Article  CAS  PubMed  Google Scholar 

  189. Sharkey, L. K. R., Edwards, T. A. & O’Neill, A. J. ABC-F proteins mediate antibiotic resistance through ribosomal protection. mBio 7, e01975-15 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  190. Swaney, S. M., Aoki, H., Ganoza, M. C. & Shinabarger, D. L. The oxazolidinone linezolid inhibits initiation of protein synthesis in bacteria. Antimicrob. Agents Chemother. 42, 3251–3255 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Schwarz, S. et al. Mobile oxazolidinone resistance genes in Gram-positive and Gram-negative bacteria. Clin. Microbiol. Rev. https://doi.org/10.1128/CMR.00188-20 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  192. Schwarz, S. et al. Lincosamides, streptogramins, phenicols, and pleuromutilins: mode of action and mechanisms of resistance. Cold Spring Harb. Perspect. Med. 6, a027037 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Gleckman, R., Blagg, N. & Joubert, D. W. Trimethoprim: mechanisms of action, antimicrobial activity, bacterial resistance, pharmacokinetics, adverse reactions, and therapeutic indications. Pharmacother. J. Hum. Pharmacol. Drug Ther. 1, 14–19 (1981).

    Article  CAS  Google Scholar 

  194. Wróbel, A., Arciszewska, K., Maliszewski, D. & Drozdowska, D. Trimethoprim and other nonclassical antifolates an excellent template for searching modifications of dihydrofolate reductase enzyme inhibitors. J. Antibiot. 73, 5–27 (2019).

    Article  Google Scholar 

  195. Correia, S., Poeta, P., Ebraud, M. H., Luis Capelo, J. & Igrejas, G. Mechanisms of quinolone action and resistance: where do we stand? J. Med. Microbiol 66, 551–559 (2017).

    Article  CAS  PubMed  Google Scholar 

  196. Floss, H. G. & Yu, T.-W. Rifamycin mode of action, resistance, and biosynthesis. Chem. Rev. 105, 621–632 (2005).

    Article  CAS  PubMed  Google Scholar 

  197. Beyer, D. & Pepper, K. The streptogramin antibiotics: update on their mechanism of action. Expert Opin. Investig. Drugs 7, 591–599 (1998).

    Article  CAS  PubMed  Google Scholar 

  198. Sköld, O. Sulfonamide resistance: mechanisms and trends. Drug Resist. Update 3, 155–160 (2000).

    Article  Google Scholar 

  199. Markley, J. L. & Wencewicz, T. A. Tetracycline-inactivating enzymes. Front. Microbiol. 9, 1058 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  200. De Pascale, G. & Wright, G. D. Antibiotic resistance by enzyme inactivation: from mechanisms to solutions. ChemBioChem 11, 1325–1334 (2010).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  202. Lambert, P. A. Bacterial resistance to antibiotics: modified target sites. Adv. Drug Deliv. Rev. 57, 1471–1485 (2005).

    Article  CAS  PubMed  Google Scholar 

  203. Then, R. L. Mechanisms of resistance to trimethoprim, the sulfonamides, and trimethoprim-sulfamethoxazole. Clin. Infect. Dis. 4, 261–269 (1982).

    Article  CAS  Google Scholar 

  204. Webber, M. A. & Piddock, L. J. V. The importance of efflux pumps in bacterial antibiotic resistance. J. Antimicrob. Chemother. 51, 9–11 (2003).

    Article  CAS  PubMed  Google Scholar 

  205. Wilson, D. N., Hauryliuk, V., Atkinson, G. C. & O’Neill, A. J. Target protection as a key antibiotic resistance mechanism. Nat. Rev. Microbiol. 18, 637–648 (2020).

    Article  CAS  PubMed  Google Scholar 

  206. Murakami, S., Nakashima, R., Yamashita, E. & Yamaguchi, A. Crystal structure of bacterial multidrug efflux transporter AcrB. Nature 419, 587–593 (2002).

    Article  ADS  CAS  PubMed  Google Scholar 

  207. Kim, J.-S. et al. Structure of the tripartite multidrug efflux pump AcrAB-TolC suggests an alternative assembly mode. Mol. Cell 38, 180–186 (2015).

    Article  Google Scholar 

  208. Zwama, M. & Yamaguchi, A. Molecular mechanisms of AcrB-mediated multidrug export. Res. Microbiol. 169, 372–383 (2018).

    Article  CAS  PubMed  Google Scholar 

  209. Cha, H., Müller, R. T. & Pos, K. M. Switch-loop flexibility affects transport of large drugs by the promiscuous AcrB multidrug efflux transporter. Antimicrob. Agents Chemother. 58, 4767–4772 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  210. Oswald, C., Tam, H.-K. & Pos, K. M. Transport of lipophilic carboxylates is mediated by transmembrane helix 2 in multidrug transporter AcrB. Nat. Commun. 7, 13819 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  211. Fischer, N. & Kandt, C. Porter domain opening and closing motions in the multi-drug efflux transporter AcrB. Biochim. Biophys. Acta Biomembr. 1828, 632–641 (2013).

    Article  CAS  Google Scholar 

  212. Rousset, F. et al. The impact of genetic diversity on gene essentiality within the Escherichia coli species. Nat. Microbiol. 6, 301–312 (2021).

    Article  CAS  PubMed  Google Scholar 

  213. Ciofu, O., Moser, C., Jensen, P. Ø. & Høiby, N. Tolerance and resistance of microbial biofilms. Nat. Rev. Microbiol. https://doi.org/10.1038/s41579-022-00682-4 (2022).

    Article  PubMed  Google Scholar 

  214. Stewart, P. S. et al. Conceptual model of biofilm antibiotic tolerance that integrates phenomena of diffusion, metabolism, gene expression, and physiology. J. Bacteriol. 201, e00307-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  215. Claessen, D. & Errington, J. Cell wall deficiency as a coping strategy for stress. Trends Microbiol. 27, 1025–1033 (2019).

    Article  CAS  PubMed  Google Scholar 

  216. Monahan, L. G. et al. Rapid conversion of Pseudomonas aeruginosa to a spherical cell morphotype facilitates tolerance to carbapenems and penicillins but increases susceptibility to antimicrobial peptides. Antimicrob. Agents Chemother. 58, 1956–1962 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  217. Balaban, N. Q. et al. Definitions and guidelines for research on antibiotic persistence. Nat. Rev. Microbiol. 17, 441–448 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Pacios, O. et al. (p)ppGpp and its role in bacterial persistence: new challenges. Antimicrob. Agents Chemother. 64, e01283-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  219. Manuse, S. et al. Bacterial persisters are a stochastically formed subpopulation of low-energy cells. PLoS Biol. 19, e3001194 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Shan, Y. et al. ATP-dependent persister formation in Escherichia coli. mBio 8, e02267-16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  221. Windels, E. M., Michiels, J. E., Van den Bergh, B., Fauvart, M. & Michiels, J. Antibiotics: combatting tolerance to stop resistance. mBio 10, e02095-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  222. Levin-Reisman, I. et al. Antibiotic tolerance facilitates the evolution of resistance. Science 355, 826–830 (2017).

    Article  ADS  CAS  PubMed  Google Scholar 

  223. Saw, H. T. H., Webber, M. A., Mushtaq, S., Woodford, N. & Piddock, L. J. V. Inactivation or inhibition of AcrAB-TolC increases resistance of carbapenemase-producing Enterobacteriaceae to carbapenems. J. Antimicrob. Chemother. 71, 1510–1519 (2016).

    Article  CAS  PubMed  Google Scholar 

  224. Ricci, V., Tzakas, P., Buckley, A., Coldham, N. C. & Piddock, L. J. V. Ciprofloxacin-resistant Salmonella enterica serovar Typhimurium strains are difficult to select in the absence of AcrB and TolC. Antimicrob. Agents Chemother. 50, 38–42 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Papkou, A., Hedge, J., Kapel, N., Young, B. & MacLean, R. C. Efflux pump activity potentiates the evolution of antibiotic resistance across S. aureus isolates. Nat. Commun. 11, 3970 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. El Meouche, I. & Dunlop, M. J. Heterogeneity in efflux pump expression predisposes antibiotic-resistant cells to mutation. Science 362, 686–690 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  227. Nolivos, S. et al. Role of AcrAB-TolC multidrug efflux pump in drug-resistance acquisition by plasmid transfer. Science 364, 778–782 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  228. Buckner, M. M. C. et al. Clinically relevant plasmid-host interactions indicate that transcriptional and not genomic modifications ameliorate fitness costs of Klebsiella pneumoniae carbapenemase-carrying plasmids. mBio 9, e02303-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  229. Zhou, Z. et al. The EnteroBase user’s guide, with case studies on Salmonella transmissions, Yersinia pestis phylogeny, and Escherichia core genomic diversity. Genome Res. 30, 138–152 (2020).

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  230. Dunn, S. J., Connor, C. & McNally, A. The evolution and transmission of multi-drug resistant Escherichia coli and Klebsiella pneumoniae: the complexity of clones and plasmids. Curr. Opin. Microbiol. 51, 51–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  231. Weber, R. E. et al. Genome-wide association studies for the detection of genetic variants associated with daptomycin and ceftaroline resistance in Staphylococcus aureus. Front. Microbiol. 12, 639660 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  232. Scribner, M. R., Santos-Lopez, A., Marshall, C. W., Deitrick, C. & Cooper, V. S. Parallel evolution of tobramycin resistance across species and environments. mBio 11, e00932-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  233. Yasir, M. et al. TraDIS-Xpress: a high-resolution whole-genome assay identifies novel mechanisms of triclosan action and resistance. Genome Res. 30, 239–249 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Jana, B. et al. The secondary resistome of multidrug-resistant Klebsiella pneumoniae. Sci. Rep. 7, 42483 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

J.M.A.B., E.M.D., E.T., P.S., M.S.G., I.A. and M.A.W. researched data for the article. J.M.A.B. and M.A.W. contributed substantially to discussion of the content. J.M.A.B., E.M.D., E.T., P.S., M.S.G., I.A. and M.A.W. wrote the article. J.M.A.B., E.M.D., E.T., P.S. and M.A.W. reviewed and/or edited the manuscript before submission.

Corresponding authors

Correspondence to Mark A. Webber or Jessica M. A. Blair.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Microbiology 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.

Related links

Beta Lactamase Database: http://www.bldb.eu

Glossary

β-Lactamase

Enzymes produced by bacteria that degrade β-lactam antibiotics.

Epistasis

Where a mutation can exert a phenotypic effect but only in concert with other genes, making the impact conditional on the genetic background of where it occurs.

Horizontal gene transfer

The movement of genetic information between bacterial cells.

Insertion sequences

Small pieces of DNA that encode their own recombination machinery and can move within or between genomes.

Minimum inhibitory concentration

(MIC). The lowest concentration of antibiotic that prevents growth of bacteria.

Topoisomerases

Essential enzymes involved in DNA replication.

Two-component system

A system that allows bacteria to respond to specific environmental stimuli. Usually, it consists of a membrane-bound histidine kinase that senses the stimuli and activates a response regulator that alters the expression of relevant genes.

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

Darby, E.M., Trampari, E., Siasat, P. et al. Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol 21, 280–295 (2023). https://doi.org/10.1038/s41579-022-00820-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-022-00820-y

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