1887

Abstract

The incidence of multidrug-resistant bacteria is increasing globally, with efflux pumps being a fundamental platform limiting drug access and synergizing with other mechanisms of resistance. Increased expression of efflux pumps is a key feature of most cells that are resistant to multiple antibiotics. Whilst expression of efflux genes can confer benefits, production of complex efflux systems is energetically costly and the expression of efflux is highly regulated, with cells balancing benefits against costs. This study used TraDIS-, a genome-wide transposon mutagenesis technology, to identify genes in and Typhimurium involved in drug efflux and its regulation. We exposed mutant libraries to the canonical efflux substrate acriflavine in the presence and absence of the efflux inhibitor phenylalanine-arginine β-naphthylamide. Comparisons between conditions identified efflux-specific and drug-specific responses. Known efflux-associated genes were easily identified, including , , , and , confirming the specificity of the response. Further genes encoding cell envelope maintenance enzymes and products involved with stringent response activation, DNA housekeeping, respiration and glutathione biosynthesis were also identified as affecting efflux activity in both species. This demonstrates the deep relationship between efflux regulation and other cellular regulatory networks. We identified a conserved set of pathways crucial for efflux activity in these experimental conditions, which expands the list of genes known to impact on efflux efficacy. Responses in both species were similar and we propose that these common results represent a core set of genes likely to be relevant to efflux control across the Enterobacteriaceae.

Keyword(s): acrB , functional genomics , marA , ramA , soxS , Tn-Seq , tolC and TraDIS
Funding
This study was supported by the:
  • Biotechnology and Biological Sciences Research Council (Award BB/R012504/1)
    • Principle Award Recipient: MarkA Webber
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.001296
2023-02-06
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/micro/169/2/mic001296.html?itemId=/content/journal/micro/10.1099/mic.0.001296&mimeType=html&fmt=ahah

References

  1. Blair JMA, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJV. Molecular mechanisms of antibiotic resistance. Nat Rev Microbiol 2015; 13:42–51 [View Article] [PubMed]
    [Google Scholar]
  2. Du D, Wang-Kan X, Neuberger A, van Veen HW, Pos KM et al. Multidrug efflux pumps: structure, function and regulation. Nat Rev Microbiol 2018; 16:523–539 [View Article]
    [Google Scholar]
  3. Blair JMA, Richmond GE, Piddock LJV. Multidrug efflux pumps in Gram-negative bacteria and their role in antibiotic resistance. Future Microbiol 2014; 9:1165–1177 [View Article]
    [Google Scholar]
  4. Li XZ, Ma D, Livermore DM, Nikaido H. Role of efflux pump(s) in intrinsic resistance of Pseudomonas aeruginosa: active efflux as a contributing factor to beta-lactam resistance. Antimicrob Agents Chemother 1994; 38:1742–1752 [View Article]
    [Google Scholar]
  5. Bailey AM, Ivens A, Kingsley R, Cottell JL, Wain J et al. RamA, a member of the AraC/XylS family, influences both virulence and efflux in Salmonella enterica serovar Typhimurium. J Bacteriol 2010; 192:1607–1616 [View Article]
    [Google Scholar]
  6. Piddock LJV. Multidrug-resistance efflux pumps - not just for resistance. Nat Rev Microbiol 2006; 4:629–636 [View Article] [PubMed]
    [Google Scholar]
  7. Kern WV, Oethinger M, Jellen-Ritter AS, Levy SB. Non-target gene mutations in the development of fluoroquinolone resistance in Escherichia coli. Antimicrob Agents Chemother 2000; 44:814–820 [View Article] [PubMed]
    [Google Scholar]
  8. Oethinger M, Kern WV, Jellen-Ritter AS, McMurry LM, Levy SB. Ineffectiveness of topoisomerase mutations in mediating clinically significant fluoroquinolone resistance in Escherichia coli in the absence of the AcrAB efflux pump. Antimicrob Agents Chemother 2000; 44:10–13 [View Article] [PubMed]
    [Google Scholar]
  9. Piddock LJV. Clinically relevant chromosomally encoded multidrug resistance efflux pumps in bacteria. Clin Microbiol Rev 2006; 19:382–402 [View Article] [PubMed]
    [Google Scholar]
  10. Gallegos MT, Schleif R, Bairoch A, Hofmann K, Ramos JL. Arac/XylS family of transcriptional regulators. Microbiol Mol Biol Rev 1997; 61:393–410 [View Article]
    [Google Scholar]
  11. Holden ER, Webber MA. MarA, rama and soxs as mediators of the stress response: survival at a cost. Front Microbiol 2020; 11:828
    [Google Scholar]
  12. Okusu H, Ma D, Nikaido H. AcrAB efflux pump plays a major role in the antibiotic resistance phenotype of Escherichia coli multiple-antibiotic-resistance (Mar) mutants. J Bacteriol 1996; 178:306–308 [View Article] [PubMed]
    [Google Scholar]
  13. Sun Y, Dai M, Hao H, Wang Y, Huang L et al. The role of RamA on the development of ciprofloxacin resistance in Salmonella enterica serovar Typhimurium. PLoS One 2011; 6:e23471 [View Article]
    [Google Scholar]
  14. Webber MA, Piddock LJ. Absence of mutations in marRAB or soxRS in acrB-overexpressing fluoroquinolone-resistant clinical and veterinary isolates of Escherichia coli. Antimicrob Agents Chemother 2001; 45:1550–1552 [View Article] [PubMed]
    [Google Scholar]
  15. Webber MA, Bailey AM, Blair JMA, Morgan E, Stevens MP et al. The global consequence of disruption of the AcrAB-TolC efflux pump in Salmonella enterica includes reduced expression of SPI-1 and other attributes required to infect the host. J Bacteriol 2009; 191:4276–4285 [View Article] [PubMed]
    [Google Scholar]
  16. Yasir M, Turner AK, Bastkowski S, Baker D, Page AJ et al. TraDIS-Xpress: a high-resolution whole-genome assay identifies novel mechanisms of triclosan action and resistance. Genome Res 2020; 30:239–249 [View Article]
    [Google Scholar]
  17. Thomson NM, Turner AK, Yasir M, Bastkowski S, Lott M et al. A whole-genome assay identifies four principal gene functions that confer tolerance of meropenem stress upon Escherichia coli. Front Antibiot 2022; 1: [View Article]
    [Google Scholar]
  18. Turner AK, Eckert SE, Turner DJ, Yasir M, Webber MA et al. A whole-genome screen identifies Salmonella enterica serovar Typhi genes involved in fluoroquinolone susceptibility. J Antimicrob Chemother 2020; 75:2516–2525 [View Article] [PubMed]
    [Google Scholar]
  19. Turner AK, Yasir M, Bastkowski S, Telatin A, Page AJ et al. A genome-wide analysis of Escherichia coli responses to fosfomycin using TraDIS-Xpress reveals novel roles for phosphonate degradation and phosphate transport systems. J Antimicrob Chemother 2020; 75:3144–3151 [View Article] [PubMed]
    [Google Scholar]
  20. Turner AK, Yasir M, Bastkowski S, Telatin A, Page A et al. Chemical biology-whole genome engineering datasets predict new antibacterial combinations. Microb Genom 2021; 7:000718 [View Article]
    [Google Scholar]
  21. Holden ER, Yasir M, Turner AK, Charles IG, Webber MA. Comparison of the genetic basis of biofilm formation between Salmonella Typhimurium and Escherichia coli. Microb Genom 2022; 8: [View Article] [PubMed]
    [Google Scholar]
  22. Holden ER, Yasir M, Turner AK, Wain J, Charles IG et al. Massively parallel transposon mutagenesis identifies temporally essential genes for biofilm formation in Escherichia coli. Microb Genom 2021; 7:000673 [View Article]
    [Google Scholar]
  23. Nakamura H. Genetic determination of resistance to acriflavine, phenethyl alcohol, and sodium dodecyl sulfate in Escherichia coli. J Bacteriol 1968; 96:987–996 [View Article]
    [Google Scholar]
  24. Trampari E, Holden ER, Wickham GJ, Ravi A, Martins L de O et al. Exposure of Salmonella biofilms to antibiotic concentrations rapidly selects resistance with collateral tradeoffs. NPJ Biofilms Microbiomes 2021; 7:3 [View Article] [PubMed]
    [Google Scholar]
  25. Barquist L, Mayho M, Cummins C, Cain AK, Boinett CJ et al. The TraDIS toolkit: sequencing and analysis for dense transposon mutant libraries. Bioinformatics 2016; 32:1109–1111 [View Article]
    [Google Scholar]
  26. Page AJ, Bastkowski S, Yasir M, Turner AK, Le Viet T et al. AlbaTraDIS: comparative analysis of large datasets from parallel transposon mutagenesis experiments. Bioinformatics 2019 [View Article]
    [Google Scholar]
  27. Baba T, Ara T, Hasegawa M, Takai Y, Okumura Y et al. Construction of Escherichia coli K-12 in-frame, single-gene knockout mutants: the Keio collection. Mol Syst Biol 2006; 2:2006.0008 [View Article]
    [Google Scholar]
  28. Lee DJ, Bingle LEH, Heurlier K, Pallen MJ, Penn CW et al. Gene doctoring: a method for recombineering in laboratory and pathogenic Escherichia coli strains. BMC Microbiol 2009; 9:252 [View Article]
    [Google Scholar]
  29. Thomson NM, Zhang C, Trampari E, Pallen MJ. Creation of golden gate constructs for gene doctoring. BMC Biotechnol 2020; 20:54 [View Article]
    [Google Scholar]
  30. Stevenson G, Neal B, Liu D, Hobbs M, Packer NH et al. Structure of the O antigen of Escherichia coli K-12 and the sequence of its rfb gene cluster. J Bacteriol 1994; 176:4144–4156 [View Article] [PubMed]
    [Google Scholar]
  31. Kalman M, Gentry DR, Cashel M. Characterization of the Escherichia coli K12 gltS glutamate permease gene. Mol Gen Genet 1991; 225:379–386 [View Article] [PubMed]
    [Google Scholar]
  32. Sá-Pessoa J, Paiva S, Ribas D, Silva IJ, Viegas SC et al. SATP (YaaH), a succinate-acetate transporter protein in Escherichia coli. Biochem J 2013; 454:585–595 [View Article] [PubMed]
    [Google Scholar]
  33. Lang S, Cressatti M, Mendoza KE, Coumoundouros CN, Plater SM et al. YehZYXW of Escherichia coli Is a low-affinity, non-osmoregulatory betaine-specific ABC transporter. Biochemistry 2015; 54:5735–5747 [View Article]
    [Google Scholar]
  34. Moussatova A, Kandt C, O’Mara ML, Tieleman DP. ATP-binding cassette transporters in Escherichia coli. Biochim Biophys Acta 2008; 1778:1757–1771 [View Article]
    [Google Scholar]
  35. Villagra NA, Hidalgo AA, Santiviago CA, Saavedra CP, Mora GC. SmvA, and not AcrB, is the major efflux pump for acriflavine and related compounds in Salmonella enterica serovar Typhimurium. J Antimicrob Chemother 2008; 62:1273–1276 [View Article] [PubMed]
    [Google Scholar]
  36. Darby EM, Trampari E, Siasat P, Gaya MS, Alav I et al. Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol 2022 [View Article] [PubMed]
    [Google Scholar]
  37. Brown L, Gentry D, Elliott T, Cashel M. DksA affects ppGpp induction of RpoS at a translational level. J Bacteriol 2002; 184:4455–4465 [View Article] [PubMed]
    [Google Scholar]
  38. Whittle EE, McNeil HE, Trampari E, Webber M, Overton TW et al. Efflux impacts intracellular accumulation only in actively growing bacterial cells. mBio 2021; 12:e0260821 [View Article]
    [Google Scholar]
  39. Wang J, Cao L, Yang X, Wu Q, Lu L et al. Transcriptional analysis reveals the critical role of RNA polymerase-binding transcription factor, DksA, in regulating multi-drug resistance of Escherichia coli. Int J Antimicrob Agents 2018; 52:63–69 [View Article] [PubMed]
    [Google Scholar]
  40. Dong T, Schellhorn HE. Global effect of RpoS on gene expression in pathogenic Escherichia coli O157:H7 strain EDL933. BMC Genomics 2009; 10:349 [View Article] [PubMed]
    [Google Scholar]
  41. Girard ME, Gopalkrishnan S, Grace ED, Halliday JA, Gourse RL et al. DksA and ppGpp regulate the σS stress response by activating promoters for the small RNA DsrA and the anti-adapter protein IraP. J Bacteriol 2018; 200:e00463-17 [View Article]
    [Google Scholar]
  42. Bougdour A, Lelong C, Geiselmann J. Crl, a low temperature-induced protein in Escherichia coli that binds directly to the stationary phase sigma subunit of RNA polymerase. J Biol Chem 2004; 279:19540–19550 [View Article] [PubMed]
    [Google Scholar]
  43. Weatherspoon-Griffin N, Yang D, Kong W, Hua Z, Shi Y. The CpxR/CpxA two-component regulatory system up-regulates the multidrug resistance cascade to facilitate Escherichia coli resistance to a model antimicrobial peptide. J Biol Chem 2014; 289:32571–32582 [View Article] [PubMed]
    [Google Scholar]
  44. Rosner JL, Martin RG. Reduction of cellular stress by TolC-dependent efflux pumps in Escherichia coli indicated by BaeSR and CpxARP activation of spy in efflux mutants. J Bacteriol 2013; 195:1042–1050 [View Article] [PubMed]
    [Google Scholar]
  45. Bontemps-Gallo S, Bohin J-P, Lacroix J-M, Slauch JM. Osmoregulated periplasmic glucans. EcoSal Plus 2017; 7: [View Article]
    [Google Scholar]
  46. Bontemps-Gallo S, Madec E, Dondeyne J, Delrue B, Robbe-Masselot C et al. Concentration of osmoregulated periplasmic glucans (OPGs) modulates the activation level of the RcsCD RcsB phosphorelay in the phytopathogen bacteria Dickeya dadantii. Environ Microbiol 2013; 15:881–894 [View Article] [PubMed]
    [Google Scholar]
  47. Baars L, Ytterberg AJ, Drew D, Wagner S, Thilo C et al. Defining the role of the Escherichia coli chaperone SecB using comparative proteomics. J Biol Chem 2006; 281:10024–10034 [View Article] [PubMed]
    [Google Scholar]
  48. Alba BM, Gross CA. Regulation of the Escherichia coli sigma-dependent envelope stress response. Mol Microbiol 2004; 52:613–619 [View Article] [PubMed]
    [Google Scholar]
  49. Adam M, Murali B, Glenn NO, Potter SS. Epigenetic inheritance based evolution of antibiotic resistance in bacteria. BMC Evol Biol 2008; 8:52 [View Article] [PubMed]
    [Google Scholar]
  50. Motta SS, Cluzel P, Aldana M. Adaptive resistance in bacteria requires epigenetic inheritance, genetic noise, and cost of efflux pumps. PLOS ONE 2015; 10:e0118464 [View Article]
    [Google Scholar]
  51. Hughes L, Roberts W, Johnson D. The impact of DNA adenine methyltransferase knockout on the development of triclosan resistance and antibiotic cross-resistance in Escherichia coli. Access Microbiol 2021; 3:acmi000178 [View Article]
    [Google Scholar]
  52. Lobner-Olesen A, Marinus MG, Hansen FG. Role of SeqA and Dam in Escherichia coli gene expression: a global/microarray analysis. In Proceedings of the National Academy of Sciences of the United States of America 2003 pp 4672–4677
    [Google Scholar]
  53. Prieto AI, Hernández SB, Cota I, Pucciarelli MG, Orlov Y et al. Roles of the outer membrane protein AsmA of Salmonella enterica in the control of marRAB expression and invasion of epithelial cells. J Bacteriol 2009; 191:3615–3622 [View Article] [PubMed]
    [Google Scholar]
  54. Cohen NR, Ross CA, Jain S, Shapiro RS, Gutierrez A et al. A role for the bacterial GATC methylome in antibiotic stress survival. Nat Genet 2016; 48:581–586 [View Article] [PubMed]
    [Google Scholar]
  55. Peterson KR, Wertman KF, Mount DW, Marinus MG. Viability of Escherichia coli K-12 DNA adenine methylase (dam) mutants requires increased expression of specific genes in the SOS regulon. Mol Gen Genet 1985; 201:14–19 [View Article]
    [Google Scholar]
  56. Lerman LS. The structure of the DNA-acridine complex. Proc Natl Acad Sci 1963; 49:94–102 [View Article]
    [Google Scholar]
  57. Valens M, Thiel A, Boccard F. The MaoP/maoS site-specific system organizes the ori region of the E. coli chromosome into a macrodomain. PLoS Genet 2016; 12:e1006309 [View Article]
    [Google Scholar]
  58. Ruiz C, Levy SB. Regulation of acrAB expression by cellular metabolites in Escherichia coli. J Antimicrob Chemother 2014; 69:390–399 [View Article] [PubMed]
    [Google Scholar]
  59. Deacon J, Cooper RA. D-Galactonate utilisation by enteric bacteria. The catabolic pathway in Escherichia coli. FEBS Lett 1977; 77:201–205 [View Article] [PubMed]
    [Google Scholar]
  60. Shafqat J, Höög JO, Hjelmqvist L, Oppermann UC, Ibáñez C et al. An ethanol-inducible MDR ethanol dehydrogenase/acetaldehyde reductase in Escherichia coli: structural and enzymatic relationships to the eukaryotic protein forms. Eur J Biochem 1999; 263:305–311 [View Article] [PubMed]
    [Google Scholar]
  61. Brock M, Darley D, Textor S, Buckel W. 2-Methylisocitrate lyases from the bacterium Escherichia coli and the filamentous fungus Aspergillus nidulans: characterization and comparison of both enzymes. Eur J Biochem 2001; 268:3577–3586 [View Article] [PubMed]
    [Google Scholar]
  62. Harborne NR, Griffiths L, Busby SJ, Cole JA. Transcriptional control, translation and function of the products of the five open reading frames of the Escherichia coli nir operon. Mol Microbiol 1992; 6:2805–2813 [View Article] [PubMed]
    [Google Scholar]
  63. De Majumdar S, Yu J, Fookes M, McAteer SP, Llobet E et al. Elucidation of the RamA regulon in Klebsiella pneumoniae reveals a role in LPS regulation. PLOS Pathog 2015; 11:e1004627 [View Article]
    [Google Scholar]
  64. Alteri CJ, Mobley HLT. Escherichia coli physiology and metabolism dictates adaptation to diverse host microenvironments. Curr Opin Microbiol 2012; 15:3–9 [View Article] [PubMed]
    [Google Scholar]
  65. Fuchs JA, Warner HR. Isolation of an Escherichia coli mutant deficient in glutathione synthesis. J Bacteriol 1975; 124:140–148 [View Article] [PubMed]
    [Google Scholar]
  66. Niehaus TD, Elbadawi-Sidhu M, de Crécy-Lagard V, Fiehn O, Hanson AD. Discovery of a widespread prokaryotic 5-oxoprolinase that was hiding in plain sight. J Biol Chem 2017; 292:16360–16367 [View Article] [PubMed]
    [Google Scholar]
  67. Song Y, Zhou Z, Gu J, Yang J, Deng J. Reducing the periplasmic glutathione content makes Escherichia coli resistant to trimethoprim and other antimicrobial drugs. Microbiol Spectr 2021; 9:e0074321 [View Article]
    [Google Scholar]
  68. Fàbrega A, Ballesté-Delpierre C, Vila J. Differential impact of ramRA mutations on both ramA transcription and decreased antimicrobial susceptibility in Salmonella Typhimurium. J Antimicrob Chemother 2016; 71:617–624 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.001296
Loading
/content/journal/micro/10.1099/mic.0.001296
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error