1887

Abstract

The gold standard method for the creation of gene deletions in is homologous recombination using allelic exchange plasmids with a temperature-sensitive origin of replication. A knockout vector that contains regions of homology is first integrated into the chromosome of by a single crossover event selected for at high temperatures (non-permissive for plasmid replication) and antibiotic selection. Next, the second crossover event is encouraged by growth without antibiotic selection at low temperature, leading at a certain frequency to the excision of the plasmid and the deletion of the gene of interest. To detect or encourage plasmid loss, either a beta-galactosidase screening method or, more typically, a counterselection step is used. We present here the adaptation of the counter-selectable marker *, coding for a mutated subunit of the phenylalanine tRNA synthetase, for use in The PheS* protein variant allows for the incorporation of the toxic phenylalanine amino acid analogue -chlorophenylalanine (PCPA) into proteins and the addition of 20–40 mM PCPA to rich media leads to drastic growth reduction for and supplementing chemically defined medium with 2.5–5 mM PCPA leads to complete growth inhibition. Using the new allelic exchange plasmid pIMAY*, we delete the magnesium transporter gene in USA300 LAC* () and RN4220 () and demonstrate that cobalt toxicity in is mainly mediated by the presence of MgtE. This new plasmid will aid the efficient and easy creation of gene knockouts in .

  • This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.000791
2019-05-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/micro/165/5/572.html?itemId=/content/journal/micro/10.1099/mic.0.000791&mimeType=html&fmt=ahah

References

  1. Thammavongsa V, Kim HK, Missiakas D, Schneewind O. Staphylococcal manipulation of host immune responses. Nat Rev Microbiol 2015; 13:529–543 [View Article][PubMed]
    [Google Scholar]
  2. Chambers HF, Deleo FR. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat Rev Microbiol 2009; 7:629–641 [View Article][PubMed]
    [Google Scholar]
  3. Arnaud M, Chastanet A, Débarbouillé M. New vector for efficient allelic replacement in naturally nontransformable, low-GC-content, gram-positive bacteria. Appl Environ Microbiol 2004; 70:6887–6891 [View Article][PubMed]
    [Google Scholar]
  4. Bae T, Schneewind O. Allelic replacement in Staphylococcus aureus with inducible counter-selection. Plasmid 2006; 55:58–63 [View Article][PubMed]
    [Google Scholar]
  5. Brückner R. Gene replacement in Staphylococcus carnosus and Staphylococcus xylosus . FEMS Microbiol Lett 1997; 151:1–8 [View Article][PubMed]
    [Google Scholar]
  6. Geiger T, Francois P, Liebeke M, Fraunholz M, Goerke C et al. The stringent response of Staphylococcus aureus and its impact on survival after phagocytosis through the induction of intracellular PSMs expression. PLoS Pathog 2012; 8:e1003016 [View Article][PubMed]
    [Google Scholar]
  7. Monk IR, Shah IM, Xu M, Tan MW, Foster TJ. Transforming the untransformable: application of direct transformation to manipulate genetically Staphylococcus aureus and Staphylococcus epidermidis . mBio 2012; 3: [View Article][PubMed]
    [Google Scholar]
  8. D'Elia MA, Pereira MP, Chung YS, Zhao W, Chau A et al. Lesions in teichoic acid biosynthesis in Staphylococcus aureus lead to a lethal gain of function in the otherwise dispensable pathway. J Bacteriol 2006; 188:4183–4189 [View Article][PubMed]
    [Google Scholar]
  9. Redder P, Linder P. New range of vectors with a stringent 5-fluoroorotic acid-based counterselection system for generating mutants by allelic replacement in Staphylococcus aureus . Appl Environ Microbiol 2012; 78:3846–3854 [View Article][PubMed]
    [Google Scholar]
  10. Chen W, Zhang Y, Yeo WS, Bae T, Ji Q. Rapid and Efficient Genome Editing in Staphylococcus aureus by Using an Engineered CRISPR/Cas9 System. J Am Chem Soc 2017; 139:3790–3795 [View Article][PubMed]
    [Google Scholar]
  11. Liu Q, Jiang Y, Shao L, Yang P, Sun B et al. CRISPR/Cas9-based efficient genome editing in Staphylococcus aureus . Acta Biochim Biophys Sin 2017; 49:764–770 [View Article][PubMed]
    [Google Scholar]
  12. Penewit K, Holmes EA, McLean K, Ren M, Waalkes A et al. Efficient and scalable precision genome editing in Staphylococcus aureus through conditional recombineering and CRISPR/Cas9-mediated counterselection. mBio 2018; 9: [View Article][PubMed]
    [Google Scholar]
  13. Yao J, Zhong J, Fang Y, Geisinger E, Novick RP et al. Use of targetrons to disrupt essential and nonessential genes in Staphylococcus aureus reveals temperature sensitivity of Ll.LtrB group II intron splicing. RNA 2006; 12:1271–1281 [View Article][PubMed]
    [Google Scholar]
  14. Kato F, Sugai M. A simple method of markerless gene deletion in Staphylococcus aureus . J Microbiol Methods 2011; 87:76–81 [View Article][PubMed]
    [Google Scholar]
  15. Sun F, Cho H, Jeong DW, Li C, He C et al. Aureusimines in Staphylococcus aureus are not involved in virulence. PLoS One 2010; 5:e15703 [View Article][PubMed]
    [Google Scholar]
  16. Leibig M, Krismer B, Kolb M, Friede A, Götz F et al. Marker removal in staphylococci via Cre recombinase and different lox sites. Appl Environ Microbiol 2008; 74:1316–1323 [View Article][PubMed]
    [Google Scholar]
  17. Pagels M, Fuchs S, Pané-Farré J, Kohler C, Menschner L et al. Redox sensing by a Rex-family repressor is involved in the regulation of anaerobic gene expression in Staphylococcus aureus . Mol Microbiol 2010; 76:1142–1161 [View Article][PubMed]
    [Google Scholar]
  18. Monk IR, Tree JJ, Howden BP, Stinear TP, Foster TJ. Complete bypass of restriction systems for major Staphylococcus aureus lineages. mBio 2015; 6:e0030800315 [View Article][PubMed]
    [Google Scholar]
  19. Ibba M, Kast P, Hennecke H. Substrate specificity is determined by amino acid binding pocket size in Escherichia coli phenylalanyl-tRNA synthetase. Biochemistry 1994; 33:7107–7112 [View Article][PubMed]
    [Google Scholar]
  20. Xie Z, Okinaga T, Qi F, Zhang Z, Merritt J. Cloning-independent and counterselectable markerless mutagenesis system in Streptococcus mutans . Appl Environ Microbiol 2011; 77:8025–8033 [View Article][PubMed]
    [Google Scholar]
  21. Gurung I, Berry JL, Hall AMJ, Pelicic V. Cloning-independent markerless gene editing in Streptococcus sanguinis: novel insights in type IV pilus biology. Nucleic Acids Res 2017; 45:e40 [View Article][PubMed]
    [Google Scholar]
  22. Kristich CJ, Chandler JR, Dunny GM. Development of a host-genotype-independent counterselectable marker and a high-frequency conjugative delivery system and their use in genetic analysis of Enterococcus faecalis . Plasmid 2007; 57:131–144 [View Article][PubMed]
    [Google Scholar]
  23. Kino Y, Nakayama-Imaohji H, Fujita M, Tada A, Yoneda S et al. Counterselection employing mutated pheS for markerless genetic deletion in Bacteroides species. Anaerobe 2016; 42:81–88 [View Article][PubMed]
    [Google Scholar]
  24. Zhou C, Shi L, Ye B, Feng H, Zhang J et al. pheS (*), an effective host-genotype-independent counter-selectable marker for marker-free chromosome deletion in Bacillus amyloliquefaciens . Appl Microbiol Biotechnol 2017; 101:217–227 [View Article][PubMed]
    [Google Scholar]
  25. Wang YC, Yuan LS, Tao HX, Jiang W, Liu CJ. pheS* as a counter-selectable marker for marker-free genetic manipulations in Bacillus anthracis . J Microbiol Methods 2018; 151:35–38 [View Article][PubMed]
    [Google Scholar]
  26. Geissendörfer M, Hillen W. Regulated expression of heterologous genes in Bacillus subtilis using the Tn10 encoded tet regulatory elements. Appl Microbiol Biotechnol 1990; 33:657–663 [View Article][PubMed]
    [Google Scholar]
  27. Helle L, Kull M, Mayer S, Marincola G, Zelder ME et al. Vectors for improved Tet repressor-dependent gradual gene induction or silencing in Staphylococcus aureus . Microbiology 2011; 157:3314–3323 [View Article][PubMed]
    [Google Scholar]
  28. Payandeh J, Pfoh R, Pai EF. The structure and regulation of magnesium selective ion channels. Biochim Biophys Acta 2013; 1828:2778–2792 [View Article][PubMed]
    [Google Scholar]
  29. Tomita A, Zhang M, Jin F, Zhuang W, Takeda H et al. ATP-dependent modulation of MgtE in Mg2+ homeostasis. Nat Commun 2017; 8:148 [View Article][PubMed]
    [Google Scholar]
  30. Schuster CF, Bellows LE, Tosi T, Campeotto I, Corrigan RM et al. The second messenger c-di-AMP inhibits the osmolyte uptake system OpuC in Staphylococcus aureus . Sci Signal 2016; 9:ra81 [View Article][PubMed]
    [Google Scholar]
  31. Price MN, Arkin AP. PaperBLAST: Text Mining Papers for Information about Homologs. mSystems 2017; 2: [View Article][PubMed]
    [Google Scholar]
  32. Armitano J, Redder P, Guimarães VA, Linder P. An Essential Factor for High Mg2+ Tolerance of Staphylococcus aureus . Front Microbiol 2016; 7:7 [View Article][PubMed]
    [Google Scholar]
  33. Nelson DL, Kennedy EP. Magnesium transport in Escherichia coli. Inhibition by cobaltous ion. J Biol Chem 1971; 246:3042–3049[PubMed]
    [Google Scholar]
  34. Snavely MD, Florer JB, Miller CG, Maguire ME. Magnesium transport in Salmonella typhimurium: 28Mg2+ transport by the CorA, MgtA, and MgtB systems. J Bacteriol 1989; 171:4761–4766 [View Article][PubMed]
    [Google Scholar]
  35. Hattori M, Iwase N, Furuya N, Tanaka Y, Tsukazaki T et al. Mg(2+)-dependent gating of bacterial MgtE channel underlies Mg(2+) homeostasis. EMBO J 2009; 28:3602–3612 [View Article][PubMed]
    [Google Scholar]
  36. Kreiswirth BN, Löfdahl S, Betley MJ, O'Reilly M, Schlievert PM et al. The toxic shock syndrome exotoxin structural gene is not detectably transmitted by a prophage. Nature 1983; 305:709–712 [View Article][PubMed]
    [Google Scholar]
  37. Boles BR, Thoendel M, Roth AJ, Horswill AR. Identification of genes involved in polysaccharide-independent Staphylococcus aureus biofilm formation. PLoS One 2010; 5:e10146 [View Article][PubMed]
    [Google Scholar]
  38. Schuster CF, Bertram R. Fluorescence based primer extension technique to determine transcriptional starting points and cleavage sites of RNases in vivo . J Vis Exp 2014; 92:e52134 [View Article][PubMed]
    [Google Scholar]
  39. Bowman L, Zeden MS, Schuster CF, Kaever V, Gründling A. New Insights into the Cyclic Di-adenosine Monophosphate (c-di-AMP) Degradation Pathway and the Requirement of the Cyclic Dinucleotide for Acid Stress Resistance in Staphylococcus aureus . J Biol Chem 2016; 291:26970–26986 [View Article][PubMed]
    [Google Scholar]
  40. Gründling A, Schneewind O. Genes required for glycolipid synthesis and lipoteichoic acid anchoring in Staphylococcus aureus . J Bacteriol 2007; 189:2521–2530 [View Article][PubMed]
    [Google Scholar]
  41. Zeden MS, Schuster CF, Bowman L, Zhong Q, Williams HD et al. Cyclic di-adenosine monophosphate (c-di-AMP) is required for osmotic regulation in Staphylococcus aureus but dispensable for viability in anaerobic conditions. J Biol Chem 2018; 293:3180–3200 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.000791
Loading
/content/journal/micro/10.1099/mic.0.000791
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF

Supplementary File 2

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