1887

Abstract

are generally soil-dwelling organisms that can also cause serious human infections. is one of the most common causative agents of infections and is often multidrug resistant. However, an additional 25 species within the genus have also been associated with infection. encodes six resistance nodulation division (RND) efflux pumps, the most clinically relevant class of efflux pumps for antibiotic export, but the distribution and types of RND efflux pumps across the genus is currently unknown. Sixty-four species making up the genus were searched for RND systems within their genomes. We also developed a novel method using conserved RND residues to predict the total number of RND proteins including currently undescribed RND pump proteins. The total number of RND proteins differed both within a species and across the genus. Species associated with infection tended to encode more pumps. AdeIJK/AdeXYZ was found in all searched species of and through genomic, structural and phenotypic work we show that these genes are actually homologues of the same system. This interpretation is further supported by structural analysis of the potential drug-binding determinants of the associated RND-transporters, which reveal their close similarity to each other, and distinctiveness from other RND-pumps in , such as AdeB. Therefore, we conclude that AdeIJK is the fundamental RND system for species in the genus . AdeIJK can export a broad range of antibiotics and provides crucial functions within the cell, for example lipid modulation of the cell membrane, and therefore it is likely that all require AdeIJK for survival and homeostasis. In contrast, additional RND systems, such as AdeABC and AdeFGH, were only found in a subset of that are associated with infection. By understanding the roles and mechanisms of RND efflux systems in , treatments for infections can avoid efflux-mediated resistance and improve patient outcomes.

Funding
This study was supported by the:
  • Wellcome Trust (Award 222386/Z/21/Z)
    • Principle Award Recipient: ElizabethM Darby
  • This is an open-access article distributed under the terms of the Creative Commons Attribution NonCommercial 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/mgen/10.1099/mgen.0.000964
2023-03-30
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/mgen/9/3/mgen000964.html?itemId=/content/journal/mgen/10.1099/mgen.0.000964&mimeType=html&fmt=ahah

References

  1. Baumann P. Isolation of Acinetobacter from soil and water. J Bacteriol 1968; 96:39–42 [View Article]
    [Google Scholar]
  2. Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 2018; 18:318–327 [View Article] [PubMed]
    [Google Scholar]
  3. Public Health England Laboratory surveillance of Acinetobacter spp. bacteraemia in England, Wales and Northern Ireland: 2018; 2019 https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/810717/hpr2119_acntbctr.pdf
  4. Chusri S, Chongsuvivatwong V, Rivera JI, Silpapojakul K, Singkhamanan K et al. Clinical outcomes of hospital-acquired infection with Acinetobacter nosocomialis and Acinetobacter pittii. Antimicrob Agents Chemother 2014; 58:4172–4179 [View Article]
    [Google Scholar]
  5. Kumar S, Patil PP, Singhal L, Ray P, Patil PB et al. Molecular epidemiology of carbapenem-resistant Acinetobacter baumannii isolates reveals the emergence of blaOXA-23 and blaNDM-1 encoding international clones in India. Infect Genet Evol 2019; 75:103986 [View Article]
    [Google Scholar]
  6. Hamouda A, Amyes SGB. Novel gyrA and parC point mutations in two strains of Acinetobacter baumannii resistant to ciprofloxacin. J Antimicrob Chemother 2004; 54:695–696 [View Article]
    [Google Scholar]
  7. Coyne S, Rosenfeld N, Lambert T, Courvalin P, Périchon B. Overexpression of resistance-nodulation-cell division pump AdeFGH confers multidrug resistance in Acinetobacter baumannii. Antimicrob Agents Chemother 2010; 54:4389–4393 [View Article] [PubMed]
    [Google Scholar]
  8. Yoon E-J, Courvalin P, Grillot-Courvalin C. RND-type efflux pumps in multidrug-resistant clinical isolates of Acinetobacter baumannii: major role for AdeABC overexpression and AdeRS mutations. Antimicrob Agents Chemother 2013; 57:2989–2995 [View Article]
    [Google Scholar]
  9. Darby EM, Trampari E, Siasat P, Gaya MS, Alav I et al. Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol 2022 [View Article]
    [Google Scholar]
  10. Klenotic PA, Moseng MA, Morgan CE, Yu EW. Structural and functional diversity of resistance-nodulation-cell division transporters. Chem Rev 2021; 121:5378–5416 [View Article]
    [Google Scholar]
  11. Alav I, Kobylka J, Kuth MS, Pos KM, Picard M et al. Structure, assembly, and function of tripartite efflux and type 1 secretion systems in Gram-negative bacteria. Chem Rev 2021; 121:5479–5596 [View Article]
    [Google Scholar]
  12. McNeil HE, Alav I, Torres RC, Rossiter AE, Laycock 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 2019; 15:e1008101 [View Article]
    [Google Scholar]
  13. Chen M, Shi X, Yu Z, Fan G, Serysheva II et al. In situ structure of the AcrAB-TolC efflux pump at subnanometer resolution. Structure 2022; 30:107–113 [View Article]
    [Google Scholar]
  14. Horiyama T, Yamaguchi A, Nishino K. TolC dependency of multidrug efflux systems in Salmonella enterica serovar Typhimurium. J Antimicrob Chemother 2010; 65:1372–1376 [View Article]
    [Google Scholar]
  15. Seeger MA, Schiefner A, Eicher T, Verrey F, Diederichs K et al. Structural asymmetry of AcrB trimer suggests a peristaltic pump mechanism. Science 2006; 313:1295–1298 [View Article]
    [Google Scholar]
  16. Murakami S, Nakashima R, Yamashita E, Matsumoto T, Yamaguchi A. Crystal structures of a multidrug transporter reveal a functionally rotating mechanism. Nature 2006; 443:173–179 [View Article]
    [Google Scholar]
  17. Nakashima R, Sakurai K, Yamasaki S, Hayashi K, Nagata C et al. Structural basis for the inhibition of bacterial multidrug exporters. Nature 2013; 500:102–106 [View Article] [PubMed]
    [Google Scholar]
  18. Nakashima R, Sakurai K, Yamasaki S, Nishino K, Yamaguchi A. Structures of the multidrug exporter AcrB reveal a proximal multisite drug-binding pocket. Nature 2011; 480:565–569 [View Article]
    [Google Scholar]
  19. Vargiu AV, Nikaido H. Multidrug binding properties of the AcrB efflux pump characterized by molecular dynamics simulations. Proc Natl Acad Sci 2012; 109:20637–20642 [View Article]
    [Google Scholar]
  20. Magnet S, Courvalin P, Lambert T. Resistance-nodulation-cell division-type efflux pump involved in aminoglycoside resistance in Acinetobacter baumannii strain BM4454. Antimicrob Agents Chemother 2001; 45:3375–3380 [View Article]
    [Google Scholar]
  21. Adams FG, Stroeher UH, Hassan KA, Marri S, Brown MH et al. Resistance to pentamidine is mediated by AdeAB, regulated by AdeRS, and influenced by growth conditions in Acinetobacter baumannii ATCC 17978. PLoS One 2018; 13:e0197412 [View Article]
    [Google Scholar]
  22. Chau S-L, Chu Y-W, Houang ETS. Novel resistance-nodulation-cell division efflux system AdeDE in Acinetobacter genomic DNA group 3. Antimicrob Agents Chemother 2004; 48:4054–4055 [View Article]
    [Google Scholar]
  23. Damier-Piolle L, Magnet S, Brémont S, Lambert T, Courvalin P. AdeIJK, a resistance-nodulation-cell division pump effluxing multiple antibiotics in Acinetobacter baumannii. Antimicrob Agents Chemother 2008; 52:557–562 [View Article]
    [Google Scholar]
  24. Chu YW, Chau SL, Houang ETS. Presence of active efflux systems AdeABC, AdeDE and AdeXYZ in different Acinetobacter genomic DNA groups. J Med Microbiol 2006; 55:477–478 [View Article]
    [Google Scholar]
  25. Williams CL, Neu HM, Gilbreath JJ, Michel SLJ, Zurawski DV et al. Copper resistance of the emerging pathogen Acinetobacter baumannii. Appl Environ Microbiol 2016; 82:6174–6188 [View Article]
    [Google Scholar]
  26. Srinivasan VB, Venkataramaiah M, Mondal A, Rajamohan G. Functional characterization of AbeD, an RND-type membrane transporter in antimicrobial resistance in Acinetobacter baumannii. PLoS One 2015; 10:e0141314 [View Article]
    [Google Scholar]
  27. Tipton KA, Farokhyfar M, Rather PN. Multiple roles for a novel RND-type efflux system in Acinetobacter baumannii AB5075. MicrobiologyOpen 2017; 6:e00418 [View Article]
    [Google Scholar]
  28. Subhadra B, Kim J, Kim DH, Woo K, Oh MH et al. Local repressor AcrR regulates AcrAB efflux pump required for biofilm formation and virulence in Acinetobacter nosocomialis. Front Cell Infect Microbiol 2018; 8:270 [View Article]
    [Google Scholar]
  29. Shi Y, Hua X, Xu Q, Yang Y, Zhang L et al. Mechanism of eravacycline resistance in Acinetobacter baumannii mediated by a deletion mutation in the sensor kinase adeS, leading to elevated expression of the efflux pump AdeABC. Infect Genet Evol 2020; 80:104185 [View Article]
    [Google Scholar]
  30. Lari AR, Ardebili A, Hashemi A. AdeR-AdeS mutations & overexpression of the AdeABC efflux system in ciprofloxacin-resistant Acinetobacter baumannii clinical isolates. Indian J Med Res 2018; 147:413–421 [View Article]
    [Google Scholar]
  31. Fernando DM, Xu W, Loewen PC, Zhanel GG, Kumar A. Triclosan can select for an AdeIJK-overexpressing mutant of Acinetobacter baumannii ATCC 17978 that displays reduced susceptibility to multiple antibiotics. Antimicrob Agents Chemother 2014; 58:6424–6431 [View Article]
    [Google Scholar]
  32. Coyne S, Guigon G, Courvalin P, Périchon B. Screening and quantification of the expression of antibiotic resistance genes in Acinetobacter baumannii with a microarray. Antimicrob Agents Chemother 2010; 54:333–340 [View Article]
    [Google Scholar]
  33. Lin L, Ling BD, Li XZ. Distribution of the multidrug efflux pump genes, adeABC, adeDE and adeIJK, and class 1 integron genes in multiple-antimicrobial-resistant clinical isolates of Acinetobacter baumannii-Acinetobacter calcoaceticus complex. Int J Antimicrob Agents 2009; 33:27–32 [View Article]
    [Google Scholar]
  34. Nemec A, Maixnerová M, van der Reijden TJK, van den Broek PJ, Dijkshoorn L. Relationship between the AdeABC efflux system gene content, netilmicin susceptibility and multidrug resistance in a genotypically diverse collection of Acinetobacter baumannii strains. J Antimicrob Chemother 2007; 60:483–489 [View Article]
    [Google Scholar]
  35. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol 2013; 30:772–780 [View Article]
    [Google Scholar]
  36. National Centre for Biotechnology Information BLAST: Basic Local Alignment Search Tool; 2021 https://blast.ncbi.nlm.nih.gov/Blast.cgi
  37. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 2014; 30:2068–2069 [View Article]
    [Google Scholar]
  38. Mikheenko A, Prjibelski A, Saveliev V, Antipov D, Gurevich A. Versatile genome assembly evaluation with QUAST-LG. Bioinformatics 2018; 34:i142–i150 [View Article]
    [Google Scholar]
  39. Jain C, Rodriguez-R LM, Phillippy AM, Konstantinidis KT, Aluru S. High throughput ANI analysis of 90K prokaryotic genomes reveals clear species boundaries. Nat Commun 2018; 9:5114 [View Article]
    [Google Scholar]
  40. Ondov BD, Treangen TJ, Melsted P, Mallonee AB, Bergman NH et al. Mash: fast genome and metagenome distance estimation using MinHash. Genome Biol 2016; 17:132 [View Article]
    [Google Scholar]
  41. Seeman T. ABRicate; 2020 https://github.com/tseemann/abricate
  42. R Core Team R: A language and environment for statistical computing. R Foundation for Statistical Computing 2017
    [Google Scholar]
  43. Wickham H. ggplot2: Elegant Graphics for Data Analysis; 2016 https://ggplot2.tidyverse.org
  44. Yu G. Using ggtree to visualize data on tree-like structures. Curr Protoc Bioinformatics 2020; 69:e96 [View Article]
    [Google Scholar]
  45. Wang L-G, Lam TT-Y, Xu S, Dai Z, Zhou L et al. Treeio: an R package for phylogenetic tree input and output with richly annotated and associated data. Mol Biol Evol 2020; 37:599–603 [View Article]
    [Google Scholar]
  46. Tonkin-Hill G, MacAlasdair N, Ruis C, Weimann A, Horesh G et al. Producing polished prokaryotic pangenomes with the Panaroo pipeline. Genome Biol 2020; 21:180 [View Article]
    [Google Scholar]
  47. Price MN, Dehal PS, Arkin AP. FastTree 2--approximately maximum-likelihood trees for large alignments. PLoS One 2010; 5:e9490 [View Article]
    [Google Scholar]
  48. IBM Corp IBM SPSS Statistics for Macintosh Armonk, NY: IBM Corp; 2021
    [Google Scholar]
  49. Sullivan MJ, Petty NK, Beatson SA. Easyfig: a genome comparison visualizer. Bioinformatics 2011; 27:1009–1010 [View Article]
    [Google Scholar]
  50. Croucher NJ, Page AJ, Connor TR, Delaney AJ, Keane JA et al. Rapid phylogenetic analysis of large samples of recombinant bacterial whole genome sequences using Gubbins. Nucleic Acids Res 2015; 43:e15 [View Article]
    [Google Scholar]
  51. Seeman T. Snippy; 2020 https://github.com/tseemann/snippy
  52. Hadfield J, Croucher NJ, Goater RJ, Abudahab K, Aanensen DM et al. Phandango: an interactive viewer for bacterial population genomics. Bioinformatics 2018; 34:292–293 [View Article]
    [Google Scholar]
  53. Letunic I, Bork P. Interactive Tree Of Life (iTOL) v5: an online tool for phylogenetic tree display and annotation. Nucleic Acids Res 2021; 49:W293–W296 [View Article]
    [Google Scholar]
  54. Zhang Z, Morgan CE, Bonomo RA, Yu EW, Projan SJ. Cryo-EM determination of eravacycline-bound structures of the ribosome and the multidrug efflux pump AdeJ of Acinetobacter baumannii. mBio 2021; 12: [View Article]
    [Google Scholar]
  55. Yang J, Yan R, Roy A, Xu D, Poisson J et al. The I-TASSER Suite: protein structure and function prediction. Nat Methods 2015; 12:7–8 [View Article]
    [Google Scholar]
  56. Eicher T, Cha H, Seeger MA, Brandstätter L, El-Delik J et al. Transport of drugs by the multidrug transporter AcrB involves an access and a deep binding pocket that are separated by a switch-loop. Proc Natl Acad Sci 2012; 109:5687–5692 [View Article]
    [Google Scholar]
  57. Ornik-Cha A, Wilhelm J, Kobylka J, Sjuts H, Vargiu AV et al. Structural and functional analysis of the promiscuous AcrB and AdeB efflux pumps suggests different drug binding mechanisms. Nat Commun 2021; 12:6919 [View Article]
    [Google Scholar]
  58. Lyu M, Moseng MA, Reimche JL, Holley CL, Dhulipala V et al. Cryo-EM structures of a gonococcal multidrug efflux pump illuminate a mechanism of drug recognition and resistance. mBio 2020; 11:e00996-20 [View Article]
    [Google Scholar]
  59. Robert X, Gouet P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res 2014; 42:W320–4 [View Article]
    [Google Scholar]
  60. Lucidi M, Runci F, Rampioni G, Frangipani E, Leoni L et al. New shuttle vectors for gene cloning and expression in multidrug-resistant Acinetobacter species. Antimicrob Agents Chemother 2018; 62: [View Article]
    [Google Scholar]
  61. Wijers CDM, Pham L, Menon S, Boyd KL, Noel HR et al. Identification of two variants of Acinetobacter baumannii strain ATCC 17978 with distinct genotypes and phenotypes. Infect Immun 2021; 89:e0045421 [View Article]
    [Google Scholar]
  62. Sambrook J, Russell DW. Transformation of E. coli by electroporation. CSH Protoc 2006; 2006:pdb.prot3933 [View Article]
    [Google Scholar]
  63. Plasmidsaurus Plasmidsaurus Sequencing. n.d https://www.plasmidsaurus.com/
  64. CLSI Performance Standards for Antimicrobial Susceptibility Testing. 31st ed 2020
    [Google Scholar]
  65. Andrews JM. Determination of minimum inhibitory concentrations. J Antimicrob Chemother 2001; 48 Suppl 1:5–16 [View Article]
    [Google Scholar]
  66. Hagman KE, Lucas CE, Balthazar JT, Snyder L, Nilles M et al. The MtrD protein of Neisseria gonorrhoeae is a member of the resistance/nodulation/division protein family constituting part of an efflux system. Microbiology 1997; 143:2117–2125 [View Article]
    [Google Scholar]
  67. Coyne S, Courvalin P, Périchon B. Efflux-mediated antibiotic resistance in Acinetobacter spp. Antimicrob Agents Chemother 2011; 55:947–953 [View Article]
    [Google Scholar]
  68. Roca I, Espinal P, Martí S, Vila J. First identification and characterization of an AdeABC-like efflux pump in Acinetobacter genomospecies 13TU. Antimicrob Agents Chemother 2011; 55:1285–1286 [View Article]
    [Google Scholar]
  69. Morgan CE, Glaza P, Leus IV, Trinh A, Su C-C et al. Cryoelectron microscopy structures of AdeB illuminate mechanisms of simultaneous binding and exporting of substrates. mBio 2021; 12:1–15 [View Article]
    [Google Scholar]
  70. Tam HK, Foong WE, Oswald C, Herrmann A, Zeng H et al. Allosteric drug transport mechanism of multidrug transporter AcrB. Nat Commun 2021; 12:3889 [View Article]
    [Google Scholar]
  71. Su C-C, Morgan CE, Kambakam S, Rajavel M, Scott H et al. Cryo-electron microscopy structure of an Acinetobacter baumannii multidrug efflux pump. mBio 2019; 10:e01295-19 [View Article]
    [Google Scholar]
  72. Murakami S, Nakashima R, Yamashita E, Yamaguchi A. Crystal structure of bacterial multidrug efflux transporter AcrB. Nature 2002; 419:587–593 [View Article] [PubMed]
    [Google Scholar]
  73. Johnson RM, Fais C, Parmar M, Cheruvara H, Marshall RL et al. Cryo-EM structure and molecular dynamics analysis of the fluoroquinolone resistant mutant of the AcrB transporter from Salmonella. Microorganisms 2020; 8:943 [View Article]
    [Google Scholar]
  74. Morgan CE, Zhang Z, Bonomo RA, Yu EW. An analysis of the novel fluorocycline TP-6076 bound to both the ribosome and multidrug efflux pump AdeJ from Acinetobacter baumannii. mBio 2022; 13:e0373221 [View Article]
    [Google Scholar]
  75. Yoon E-J, Balloy V, Fiette L, Chignard M, Courvalin P et al. Contribution of the Ade resistance-nodulation-cell division-type efflux pumps to fitness and pathogenesis of Acinetobacter baumannii. mBio 2016; 7:e00697-16 [View Article]
    [Google Scholar]
  76. Jerse AE, Sharma ND, Simms AN, Crow ET, Snyder LA et al. A gonococcal efflux pump system enhances bacterial survival in a female mouse model of genital tract infection. Infect Immun 2003; 71:5576–5582 [View Article]
    [Google Scholar]
  77. Buckley AM, Webber MA, Cooles S, Randall LP, La Ragione RM et al. The AcrAB-TolC efflux system of Salmonella enterica serovar Typhimurium plays a role in pathogenesis. Cell Microbiol 2006; 8:847–856 [View Article]
    [Google Scholar]
  78. Hirakata Y, Srikumar R, Poole K, Gotoh N, Suematsu T et al. Multidrug efflux systems play an important role in the invasiveness of Pseudomonas aeruginosa. J Exp Med 2002; 196:109–118 [View Article]
    [Google Scholar]
  79. Lin J, Sahin O, Michel LO, Zhang Q. Critical role of multidrug efflux pump CmeABC in bile resistance and in vivo colonization of Campylobacter jejuni. Infect Immun 2003; 71:4250–4259 [View Article]
    [Google Scholar]
  80. Bina XR, Provenzano D, Nguyen N, Bina JE. Vibrio cholerae RND family efflux systems are required for antimicrobial resistance, optimal virulence factor production, and colonization of the infant mouse small intestine. Infect Immun 2008; 76:3595–3605 [View Article]
    [Google Scholar]
  81. Kornelsen V, Kumar A. Update on multidrug resistance efflux pumps in Acinetobacter spp. Antimicrob Agents Chemother 2021; 65:e0051421 [View Article]
    [Google Scholar]
  82. Sugawara E, Nikaido H. Properties of AdeABC and AdeIJK efflux systems of Acinetobacter baumannii compared with those of the AcrAB-TolC system of Escherichia coli. Antimicrob Agents Chemother 2014; 58:7250–7257 [View Article]
    [Google Scholar]
  83. Zwama M, Yamasaki S, Nakashima R, Sakurai K, Nishino K et al. Multiple entry pathways within the efflux transporter AcrB contribute to multidrug recognition. Nat Commun 2018; 9:124 [View Article]
    [Google Scholar]
  84. Sennhauser G, Bukowska MA, Briand C, Grütter MG. Crystal structure of the multidrug exporter MexB from Pseudomonas aeruginosa. J Mol Biol 2009; 389:134–145 [View Article]
    [Google Scholar]
  85. Zwama M, Nishino K. Proximal Binding Pocket Arg717 Substitutions in Escherichia coli AcrB Cause Clinically Relevant Divergencies in Resistance Profiles. Antimicrobial agents and chemotherapy 2022; 66:e02392-21 [View Article]
    [Google Scholar]
  86. Chitsaz M, Booth L, Blyth MT, O’Mara ML, Brown MH. Multidrug Resistance in Neisseria gonorrhoeae: Identification of Functionally Important Residues in the MtrD Efflux Protein. mBio 2019; 10:e02277-19 [View Article]
    [Google Scholar]
  87. Hernández-González IL, Mateo-Estrada V, Castillo-Ramirez S. The promiscuous and highly mobile resistome of Acinetobacter baumannii. Microb Genom 2022; 8:000762 [View Article]
    [Google Scholar]
  88. Naidu V, Bartczak A, Brzoska AJ, Lewis P, Eijkelkamp BA et al. Evolution of RND efflux pumps in the development of a successful pathogen. Drug Resist Updat 2023; 66:100911 [View Article] [PubMed]
    [Google Scholar]
  89. Yoon E-J, Chabane YN, Goussard S, Snesrud E, Courvalin P et al. Contribution of resistance-nodulation-cell division efflux systems to antibiotic resistance and biofilm formation in Acinetobacter baumannii. mBio 2015; 6:e00309-15 [View Article]
    [Google Scholar]
  90. Knight DB, Rudin SD, Bonomo RA, Rather PN. Acinetobacter nosocomialis: defining the role of efflux pumps in resistance to antimicrobial therapy, surface motility, and biofilm formation. Front Microbiol 2018; 9:1902 [View Article]
    [Google Scholar]
  91. Anes J, McCusker MP, Fanning S, Martins M. The ins and outs of RND efflux pumps in Escherichia coli. Front Microbiol 2015; 6:587 [View Article]
    [Google Scholar]
  92. Zwama M, Yamaguchi A, Nishino K. Phylogenetic and functional characterisation of the Haemophilus influenzae multidrug efflux pump AcrB. Commun Biol 2019; 2:340 [View Article]
    [Google Scholar]
  93. Zulfiqar S, Shakoori AR. Molecular characterization, metal uptake and copper induced transcriptional activation of efflux determinants in copper resistant isolates of Klebsiella pneumoniae. Gene 2012; 510:32–38 [View Article]
    [Google Scholar]
  94. Ni RT, Onishi M, Mizusawa M, Kitagawa R, Kishino T et al. The role of RND-type efflux pumps in multidrug-resistant mutants of Klebsiella pneumoniae. Sci Rep 2020; 10:10876 [View Article]
    [Google Scholar]
  95. Pasqua M, Grossi M, Scinicariello S, Aussel L, Barras F et al. The MFS efflux pump EmrKY contributes to the survival of Shigella within macrophages. Sci Rep 2019; 9:2906 [View Article]
    [Google Scholar]
  96. Stothard P. The sequence manipulation suite: JavaScript programs for analyzing and formatting protein and DNA sequences. Biotechniques 2000; 28:1102, 1104 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.000964
Loading
/content/journal/mgen/10.1099/mgen.0.000964
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Supplementary material 2

PDF

Supplementary material 3

PDF

Supplementary material 4

PDF

Supplementary material 5

TEXT

Supplementary material 6

TEXT
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