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:

The bacterial epigenome

Abstract

In all domains of life, genomes contain epigenetic information superimposed over the nucleotide sequence. Epigenetic signals control DNA–protein interactions and can cause phenotypic change in the absence of mutation. A nearly universal mechanism of epigenetic signalling is DNA methylation. In bacteria, DNA methylation has roles in genome defence, chromosome replication and segregation, nucleoid organization, cell cycle control, DNA repair and regulation of transcription. In many bacterial species, DNA methylation controls reversible switching (phase variation) of gene expression, a phenomenon that generates phenotypic cell variants. The formation of epigenetic lineages enables the adaptation of bacterial populations to harsh or changing environments and modulates the interaction of pathogens with their eukaryotic hosts.

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

Access options

Buy this article

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

Fig. 1: Methylation, hemimethylation and passive demethylation of GATC sites in gammaproteobacteria.
Fig. 2: Control of bistable loci by the formation of alternative Dam methylation patterns.
Fig. 3: Formation of subpopulations controlled by Dam-dependent methylation.
Fig. 4: Mechanisms of switching and methylome diversification in phasevarions.

Similar content being viewed by others

References

  1. Holliday, R. Epigenetics: a historical overview. Epigenetics 1, 76–80 (2006).

    PubMed  Google Scholar 

  2. Henikoff, S. & Greally, J. M. Epigenetics, cellular memory and gene regulation. Curr. Biol. 26, R644–R648 (2016).

    CAS  PubMed  Google Scholar 

  3. Jones, P. A. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 13, 484–492 (2012).

    CAS  PubMed  Google Scholar 

  4. Lawrence, M., Daujat, S. & Schneider, R. Lateral thinking: How histone modifications regulate gene expression. Trends Genet. 32, 42–56 (2016).

    CAS  PubMed  Google Scholar 

  5. Marinus, M. G. Methylation of DNA in Escherichia coli and Salmonella. Cell. Mol. Biol. 782–791 (1996).

  6. Casadesus, J. & Low, D. Epigenetic gene regulation in the bacterial world. Microbiol. Mol. Biol. Rev. 70, 830–856 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Wion, D. & Casadesus, J. N6-methyl-adenine: an epigenetic signal for DNA–protein interactions. Nat. Rev. Microbiol. 4, 183–192 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Løbner-Olesen, A., Skovgaard, O. & Marinus, M. G. Dam methylation: coordinating cellular processes. Curr. Opin. Microbiol. 8, 154–160 (2005).

    PubMed  Google Scholar 

  9. Stephens, C., Reisenauer, A., Wright, R. & Shapiro, L. A cell cycle-regulated bacterial DNA methyltransferase is essential for viability. Proc. Natl Acad. Sci. USA 93, 1210–1214 (1996).

    CAS  PubMed  Google Scholar 

  10. Blyn, L. B., Braaten, B. A., White-Ziegler, C. A., Rolfson, D. H. & Low, D. A. Phase-variation of pyelonephritis-associated pili in Escherichia coli: evidence for transcriptional regulation. EMBO J. 8, 613–620 (1989). This study provides the first description of bacterial lineage formation under the control of DNA methylation.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Heithoff, D. M., Sinsheimer, R. L., Low, D. A. & Mahan, M. J. An essential role for DNA adenine methylation in bacterial virulence. Science 284, 967–970 (1999).

    CAS  PubMed  Google Scholar 

  12. Garcia-Del Portillo, F., Pucciarelli, M. G. & Casadesus, J. DNA adenine methylase mutants of Salmonella typhimurium show defects in protein secretion, cell invasion, and M cell cytotoxicity. Proc. Natl Acad. Sci. USA 96, 11578–11583 (1999).

    CAS  PubMed  Google Scholar 

  13. Marinus, M. G. & Casadesus, J. Roles of DNA adenine methylation in host-pathogen interactions: mismatch repair, transcriptional regulation, and more. FEMS Microbiol. Rev. 33, 488–503 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Sanchez-Romero, M. A., Cota, I. & Casadesus, J. DNA methylation in bacteria: from the methyl group to the methylome. Curr. Opin. Microbiol. 25, 9–16 (2015).

    CAS  PubMed  Google Scholar 

  15. Casadesus, J. & Low, D. A. Programmed heterogeneity: epigenetic mechanisms in bacteria. J. Biol. Chem. 288, 13929–13935 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Casadesús, J. & Torreblanca, J. in Epigenetic Mechanisms of Gene Regulation (eds. Russo, V. E. A., Martienssen, R. A. & Riggs, A. D.) 141–153 (Cold Spring Harbor Laboratory, 1996).

  17. Atack, J. M., Tan, A., Bakaletz, L. O., Jennings, M. P. & Seib, K. L. Phasevarions of bacterial pathogens: methylomics sheds new Light on old enemies. Trends Microbiol. 26, 715–726 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen, C. et al. Convergence of DNA methylation and phosphorothioation epigenetics in bacterial genomes. Proc. Natl Acad. Sci. USA 114, 4501–4506 (2017).

    CAS  PubMed  Google Scholar 

  19. Wang, L., Jiang, S., Deng, Z., Dedon, P. C. & Chen, S. DNA phosphorothioate modification—a new multi-functional epigenetic system in bacteria. FEMS Microbiol. Rev. 43, 109–122 (2019).

    CAS  PubMed  Google Scholar 

  20. Flusberg, B. A. et al. Direct detection of DNA methylation during single-molecule, real-time sequencing. Nat. Methods 7, 461–465 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Fang, G. et al. Genome-wide mapping of methylated adenine residues in pathogenic Escherichia coli using single-molecule real-time sequencing. Nat. Biotechnol. 30, 1232–1239 (2012).

    CAS  PubMed  Google Scholar 

  22. Clarke, J. et al. Continuous base identification for single-molecule nanopore DNA sequencing. Nat. Nanotechnol. 4, 265–270 (2009).

    CAS  PubMed  Google Scholar 

  23. Beaulaurier, J., Schadt, E. E. & Fang, G. Deciphering bacterial epigenomes using modern sequencing technologies. Nat. Rev. Genet. 20, 157–172 (2019). This review summarizes the technologies for mapping bacterial methylomes.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Blow, M. J. et al. The epigenomic landscape of prokaryotes. PLOS Genet. 12, e1005854 (2016).

    PubMed  PubMed Central  Google Scholar 

  25. Lluch-Senar, M. et al. Comprehensive methylome characterization of Mycoplasma genitalium and Mycoplasma pneumoniae at single-base resolution. PLOS Genet. 9, e1003191 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Vasu, K. & Nagaraja, V. Diverse functions of restriction-modification systems in addition to cellular defense. Microbiol. Mol. Biol. Rev. 77, 53–72 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. De Ste Croix, M. et al. Phase-variable methylation and epigenetic regulation by type I restriction-modification systems. FEMS Microbiol. Rev. 41, S3–S15 (2017).

    Google Scholar 

  28. Tan, A., Atack, J. M., Jennings, M. P. & Seib, K. L. The capricious nature of bacterial pathogens: phasevarions and vaccine development. Front. Immunol. 7, 586 (2016).

    PubMed  PubMed Central  Google Scholar 

  29. Thattai, M. & van Oudenaarden, A. Stochastic gene expression in fluctuating environments. Genetics 167, 523–530 (2004).

    PubMed  PubMed Central  Google Scholar 

  30. Kussell, E. & Leibler, S. Phenotypic diversity, population growth, and information in fluctuating environments. Science 309, 2075–2078 (2005).

    CAS  PubMed  Google Scholar 

  31. Adhikari, S. & Curtis, P. D. DNA methyltransferases and epigenetic regulation in bacteria. FEMS Microbiol. Rev. 40, 575–591 (2016).

    CAS  PubMed  Google Scholar 

  32. Mouammine, A. & Collier, J. The impact of DNA methylation in Alphaproteobacteria. Mol. Microbiol. 110, 1–10 (2018). This recent article reviews DNA methylation in alphaproteobacteria.

    CAS  PubMed  Google Scholar 

  33. Phillips, Z. N., Husna, A. U., Jennings, M. P., Seib, K. L. & Atack, J. M. Phasevarions of bacterial pathogens-phase-variable epigenetic regulators evolving from restriction-modification systems. Microbiology 165, 917–928 (2019). This recent review covers phasevarions.

    CAS  PubMed  Google Scholar 

  34. Mohapatra, S. S., Fioravanti, A. & Biondi, E. G. DNA methylation in Caulobacter and other Alphaproteobacteria during cell cycle progression. Trends Microbiol. 22, 528–535 (2014).

    CAS  PubMed  Google Scholar 

  35. Jurkowska, R. Z. & Jeltsch, A. Mechanisms and biological roles of DNA methyltransferases and DNA methylation: from past achievements to future challenges. Adv. Exp. Med. Biol. 945, 1–17 (2016).

    CAS  PubMed  Google Scholar 

  36. Cheng, X. Structure and function of DNA methyltransferases. Annu. Rev. Biophys. Biomol. Struct. 24, 293–318 (1995).

    CAS  PubMed  Google Scholar 

  37. Bheemanaik, S., Reddy, Y. V. & Rao, D. N. Structure, function and mechanism of exocyclic DNA methyltransferases. Biochem. J. 399, 177–190 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Malone, T., Blumenthal, R. M. & Cheng, X. Structure-guided analysis reveals nine sequence motifs conserved among DNA amino-methyltransferases, and suggests a catalytic mechanism for these enzymes. J. Mol. Biol. 253, 618–632 (1995).

    CAS  PubMed  Google Scholar 

  39. Bujnicki, J. M. Sequence permutations in the molecular evolution of DNA methyltransferases. BMC Evol. Biol. 2, 3 (2002).

    PubMed  PubMed Central  Google Scholar 

  40. Ershova, A. S., Rusinov, I. S., Spirin, S. A., Karyagina, A. S. & Alexeevski, A. V. Role of restriction-modification systems in prokaryotic evolution and ecology. Biochem. 80, 1373–1386 (2015).

    CAS  Google Scholar 

  41. Oliveira, P. H., Touchon, M. & Rocha, E. P. The interplay of restriction-modification systems with mobile genetic elements and their prokaryotic hosts. Nucleic Acids Res. 42, 10618–10631 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Nobusato, A., Uchiyama, I. & Kobayashi, I. Diversity of restriction-modification gene homologues in Helicobacter pylori. Gene 259, 89–98 (2000).

    CAS  PubMed  Google Scholar 

  43. Murphy, J., Mahony, J., Ainsworth, S., Nauta, A. & van Sinderen, D. Bacteriophage orphan DNA methyltransferases: insights from their bacterial origin, function, and occurrence. Appl. Env. Microbiol. 79, 7547–7555 (2013).

    CAS  Google Scholar 

  44. Broadbent, S. E., Balbontin, R., Casadesus, J., Marinus, M. G. & van der Woude, M. YhdJ, a nonessential CcrM-like DNA methyltransferase of Escherichia coli and Salmonella enterica. J. Bacteriol. 189, 4325–4327 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Murray, I. A. et al. The non-specific adenine DNA methyltransferase M.EcoGII. Nucleic Acids Res. 46, 840–848 (2018).

    CAS  PubMed  Google Scholar 

  46. Urig, S. et al. The Escherichia coli dam DNA methyltransferase modifies DNA in a highly processive reaction. J. Mol. Biol. 319, 1085–1096 (2002).

    CAS  PubMed  Google Scholar 

  47. Albu, R. F., Jurkowski, T. P. & Jeltsch, A. The Caulobacter crescentus DNA-(adenine-N6)-methyltransferase CcrM methylates DNA in a distributive manner. Nucleic Acids Res. 40, 1708–1716 (2012).

    CAS  PubMed  Google Scholar 

  48. Peterson, S. N. & Reich, N. O. GATC flanking sequences regulate Dam activity: evidence for how Dam specificity may influence pap expression. J. Mol. Biol. 355, 459–472 (2006). This study describes DNA sequence elements that influence the processivity of the Dam methyltransferase.

    CAS  PubMed  Google Scholar 

  49. Payelleville, A. et al. DNA adenine methyltransferase (Dam) overexpression impairs Photorhabdus luminescens motility and virulence. Front. Microbiol. 8, 1671 (2017).

    PubMed  PubMed Central  Google Scholar 

  50. Julio, S. M. et al. DNA adenine methylase is essential for viability and plays a role in the pathogenesis of Yersinia pseudotuberculosis and Vibrio cholerae. Infect. Immun. 69, 7610–7615 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Robinson, V. L., Oyston, P. C. & Titball, R. W. A dam mutant of Yersinia pestis is attenuated and induces protection against plague. FEMS Microbiol. Lett. 252, 251–256 (2005).

    CAS  PubMed  Google Scholar 

  52. Taylor, V. L., Titball, R. W. & Oyston, P. C. Oral immunization with a dam mutant of Yersinia pseudotuberculosis protects against plague. Microbiology 151, 1919–1926 (2005).

    CAS  PubMed  Google Scholar 

  53. Gonzalez, D. & Collier, J. DNA methylation by CcrM activates the transcription of two genes required for the division of Caulobacter crescentus. Mol. Microbiol. 88, 203–218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Falker, S., Schilling, J., Schmidt, M. A. & Heusipp, G. Overproduction of DNA adenine methyltransferase alters motility, invasion, and the lipopolysaccharide O-antigen composition of Yersinia enterocolitica. Infect. Immun. 75, 4990–4997 (2007).

    PubMed  PubMed Central  Google Scholar 

  55. Shell, S. S. et al. DNA methylation impacts gene expression and ensures hypoxic survival of Mycobacterium tuberculosis. PLOS Pathog. 9, e1003419 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Balbontin, R. et al. DNA adenine methylation regulates virulence gene expression in Salmonella enterica serovar Typhimurium. J. Bacteriol. 188, 8160–8168 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Gonzalez, D., Kozdon, J. B., McAdams, H. H., Shapiro, L. & Collier, J. The functions of DNA methylation by CcrM in Caulobacter crescentus: a global approach. Nucleic Acids Res. 42, 3720–3735 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Friso, S., Udali, S., De Santis, D. & Choi, S. W. One-carbon metabolism and epigenetics. Mol. Asp. Med. 54, 28–36 (2017).

    CAS  Google Scholar 

  59. Torreblanca, J. & Casadesus, J. DNA adenine methylase mutants of Salmonella typhimurium and a novel Dam-regulated locus. Genetics 144, 15–26 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Marinus, M. G. Recombination is essential for viability of an Escherichia coli dam (DNA adenine methyltransferase) mutant. J. Bacteriol. 182, 463–468 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Campellone, K. G. et al. Increased adherence and actin pedestal formation by dam-deficient enterohaemorrhagic Escherichia coli O157:H7. Mol. Microbiol. 63, 1468–1481 (2007).

    CAS  PubMed  Google Scholar 

  62. Lopez-Garrido, J. & Casadesus, J. Regulation of Salmonella enterica pathogenicity island 1 by DNA adenine methylation. Genetics 184, 637–649 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Herman, G. E. & Modrich, P. Escherichia coli dam methylase. Physical and catalytic properties of the homogeneous enzyme. J. Biol. Chem. 257, 2605–2612 (1982).

    CAS  PubMed  Google Scholar 

  64. Boye, E., Marinus, M. G. & Lobner-Olesen, A. Quantitation of Dam methyltransferase in Escherichia coli. J. Bacteriol. 174, 1682–1685 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Campbell, J. L. & Kleckner, N. The rate of Dam-mediated DNA adenine methylation in Escherichia coli. Gene 74, 189–190 (1988).

    CAS  PubMed  Google Scholar 

  66. Low, D. A. & Casadesus, J. Clocks and switches: bacterial gene regulation by DNA adenine methylation. Curr. Opin. Microbiol. 11, 106–112 (2008).

    CAS  PubMed  Google Scholar 

  67. Peterson, S. N. & Reich, N. O. Competitive Lrp and Dam assembly at the pap regulatory region: implications for mechanisms of epigenetic regulation. J. Mol. Biol. 383, 92–105 (2008).

    CAS  PubMed  Google Scholar 

  68. Horton, J. R., Liebert, K., Bekes, M., Jeltsch, A. & Cheng, X. Structure and substrate recognition of the Escherichia coli DNA adenine methyltransferase. J. Mol. Biol. 358, 559–570 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. van der Woude, M. W., Braaten, B. A. & Low, D. A. Evidence for global regulatory control of pilus expression in Escherichia coli by Lrp and DNA methylation: model building based on analysis of pap. Mol. Microbiol. 6, 2429–2435 (1992).

    PubMed  Google Scholar 

  70. Wallecha, A., Munster, V., Correnti, J., Chan, T. & van der Woude, M. Dam- and OxyR-dependent phase variation of agn43: essential elements and evidence for a new role of DNA methylation. J. Bacteriol. 184, 3338–3347 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Waldron, D. E., Owen, P. & Dorman, C. J. Competitive interaction of the OxyR DNA-binding protein and the Dam methylase at the antigen 43 gene regulatory region in Escherichia coli. Mol. Microbiol. 44, 509–520 (2002).

    CAS  PubMed  Google Scholar 

  72. Cota, I. et al. OxyR-dependent formation of DNA methylation patterns in OpvABOFF and OpvABON cell lineages of Salmonella enterica. Nucleic Acids Res. 44, 3595–3609 (2016).

    CAS  PubMed  Google Scholar 

  73. Brunet, Y. R., Bernard, C. S., Gavioli, M., Lloubes, R. & Cascales, E. An epigenetic switch involving overlapping Fur and DNA methylation optimizes expression of a type VI secretion gene cluster. PLOS Genet. 7, e1002205 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Jakomin, M., Chessa, D., Baumler, A. J. & Casadesus, J. Regulation of the Salmonella enterica std fimbrial operon by DNA adenine methylation, SeqA, and HdfR. J. Bacteriol. 190, 7406–7413 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Garcia-Pastor, L., Sanchez-Romero, M. A., Jakomin, M., Puerta-Fernandez, E. & Casadesus, J. Regulation of bistability in the std fimbrial operon of Salmonella enterica by DNA adenine methylation and transcription factors HdfR, StdE and StdF. Nucleic Acids Res. 47, 7929–7941 (2019).

    PubMed  PubMed Central  Google Scholar 

  76. Ardissone, S. et al. Cell cycle constraints and environmental control of local DNA hypomethylation in alpha-proteobacteria. PLOS Genet. 12, e1006499 (2016).

    PubMed  PubMed Central  Google Scholar 

  77. Ringquist, S. & Smith, C. L. The Escherichia coli chromosome contains specific, unmethylated dam and dcm sites. Proc. Natl Acad. Sci. USA 89, 4539–4543 (1992).

    CAS  PubMed  Google Scholar 

  78. Wang, M. X. & Church, G. M. A whole genome approach to in vivo DNA–protein interactions in E. coli. Nature 360, 606–610 (1992).

    CAS  PubMed  Google Scholar 

  79. Kozdon, J. B. et al. Global methylation state at base-pair resolution of the Caulobacter genome throughout the cell cycle. Proc. Natl Acad. Sci. USA 110, E4658–E4667 (2013).

    CAS  PubMed  Google Scholar 

  80. Payelleville, A. et al. The complete methylome of an entomopathogenic bacterium reveals the existence of loci with unmethylated adenines. Sci. Rep. 8, 12091 (2018).

    PubMed  PubMed Central  Google Scholar 

  81. Hale, W. B., van der Woude, M. W. & Low, D. A. Analysis of nonmethylated GATC sites in the Escherichia coli chromosome and identification of sites that are differentially methylated in response to environmental stimuli. J. Bacteriol. 176, 3438–3441 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Li, D. et al. Exocyclic carbons adjacent to the N6 of adenine are targets for oxidation by the Escherichia coli adaptive response protein AlkB. J. Am. Chem. Soc. 134, 8896–8901 (2012). This report describes active demethylation of 6mA during DNA repair.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Polaczek, P., Kwan, K. & Campbell, J. L. GATC motifs may alter the conformation of DNA depending on sequence context and N6-adenine methylation status: possible implications for DNA–protein recognition. Mol. Gen. Genet. 258, 488–493 (1998).

    CAS  PubMed  Google Scholar 

  84. Kimura, T., Asai, T., Imai, M. & Takanami, M. Methylation strongly enhances DNA bending in the replication origin region of the Escherichia coli chromosome. Mol. Gen. Genet. 219, 69–74 (1989).

    CAS  PubMed  Google Scholar 

  85. Diekmann, S. DNA methylation can enhance or induce DNA curvature. EMBO J. 6, 4213–4217 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Waldminghaus, T. & Skarstad, K. The Escherichia coli SeqA protein. Plasmid 61, 141–150 (2009).

    CAS  PubMed  Google Scholar 

  87. Sanchez-Romero, M. A. et al. Dynamic distribution of SeqA protein across the chromosome of Escherichia coli K-12. Mbio 1, e00012-10 (2010).

    PubMed  PubMed Central  Google Scholar 

  88. Camacho, E. M. & Casadesus, J. Conjugal transfer of the virulence plasmid of Salmonella enterica is regulated by the leucine-responsive regulatory protein and DNA adenine methylation. Mol. Microbiol. 44, 1589–1598 (2002).

    CAS  PubMed  Google Scholar 

  89. Camacho, E. M. & Casadesus, J. Regulation of traJ transcription in the Salmonella virulence plasmid by strand-specific DNA adenine hemimethylation. Mol Microbiol 57, 1700–1718 (2005).

    CAS  PubMed  Google Scholar 

  90. Fioravanti, A. et al. DNA binding of the cell cycle transcriptional regulator GcrA depends on N6-adenosine methylation in Caulobacter crescentus and other Alphaproteobacteria. PLOS Genet. 9, e1003541 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Roberts, D., Hoopes, B. C., McClure, W. R. & Kleckner, N. IS10 transposition is regulated by DNA adenine methylation. Cell 43, 117–130 (1985).

    CAS  PubMed  Google Scholar 

  92. Camacho, E. M. et al. Regulation of finP transcription by DNA adenine methylation in the virulence plasmid of Salmonella enterica. J. Bacteriol. 187, 5691–5699 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. van der Woude, M., Braaten, B. & Low, D. Epigenetic phase variation of the pap operon in Escherichia coli. Trends Microbiol 4, 5–9 (1996).

    PubMed  Google Scholar 

  94. Broadbent, S. E., Davies, M. R. & van der Woude, M. W. Phase variation controls expression of Salmonella lipopolysaccharide modification genes by a DNA methylation-dependent mechanism. Mol. Microbiol. 77, 337–353 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Hernday, A. D., Braaten, B. A. & Low, D. A. The mechanism by which DNA adenine methylase and PapI activate the pap epigenetic switch. Mol. Cell 12, 947–957 (2003).

    CAS  PubMed  Google Scholar 

  96. Davies, M. R., Broadbent, S. E., Harris, S. R., Thomson, N. R. & van der Woude, M. W. Horizontally acquired glycosyltransferase operons drive salmonellae lipopolysaccharide diversity. PLOS Genet. 9, e1003568 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Garcia-Pastor, L., Sanchez-Romero, M. A., Gutierrez, G., Puerta-Fernandez, E. & Casadesus, J. Formation of phenotypic lineages in Salmonella enterica by a pleiotropic fimbrial switch. PLOS Genet. 14, e1007677 (2018).

    PubMed  PubMed Central  Google Scholar 

  98. Suwandi, A. et al. Std fimbriae–fucose interaction increases Salmonella-induced intestinal inflammation and prolongs colonization. PLOS Pathog. 15, e1007915 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Weyand, N. J., Braaten, B. A., van der Woude, M., Tucker, J. & Low, D. A. The essential role of the promoter-proximal subunit of CAP in pap phase variation: Lrp- and helical phase-dependent activation of papBA transcription by CAP from -215. Mol. Microbiol. 39, 1504–1522 (2001).

    CAS  PubMed  Google Scholar 

  100. Hernday, A. D., Braaten, B. A., Broitman-Maduro, G., Engelberts, P. & Low, D. A. Regulation of the pap epigenetic switch by CpxAR: phosphorylated CpxR inhibits transition to the phase ON state by competition with Lrp. Mol. Cell 16, 537–547 (2004).

    CAS  PubMed  Google Scholar 

  101. White-Ziegler, C. A., Angus Hill, M. L., Braaten, B. A., van der Woude, M. W. & Low, D. A. Thermoregulation of Escherichia coli pap transcription: H-NS is a temperature-dependent DNA methylation blocking factor. Mol. Microbiol. 28, 1121–1137 (1998).

    CAS  PubMed  Google Scholar 

  102. Reyes-Lamothe, R. & Sherratt, D. J. The bacterial cell cycle, chromosome inheritance and cell growth. Nat. Rev. Microbiol. 17, 467–478 (2019).

    CAS  PubMed  Google Scholar 

  103. Campbell, J. L. & Kleckner, N. E. coli oric and the dnaA gene promoter are sequestered from dam methyltransferase following the passage of the chromosomal replication fork. Cell 62, 967–979 (1990).

    CAS  PubMed  Google Scholar 

  104. Cagliero, C., Grand, R. S., Jones, M. B., Jin, D. J. & O’Sullivan, J. M. Genome conformation capture reveals that the Escherichia coli chromosome is organized by replication and transcription. Nucleic Acids Res. 41, 6058–6071 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Modrich, P. Methyl-directed DNA mismatch correction. J. Biol. Chem. 264, 6597–6600 (1989).

    CAS  PubMed  Google Scholar 

  106. Cota, I., Blanc-Potard, A. B. & Casadesus, J. STM2209-STM2208 (opvAB): a phase variation locus of Salmonella enterica involved in control of O-antigen chain length. PLOS ONE 7, e36863 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Cota, I. et al. Epigenetic control of Salmonella enterica O-antigen chain length: a tradeoff between virulence and bacteriophage resistance. PLOS Genet. 11, e1005667 (2015).

    PubMed  PubMed Central  Google Scholar 

  108. Chessa, D., Winter, M. G., Jakomin, M. & Baumler, A. J. Salmonella enterica serotype Typhimurium Std fimbriae bind terminal alpha(1,2)fucose residues in the cecal mucosa. Mol. Microbiol. 71, 864–875 (2009).

    CAS  PubMed  Google Scholar 

  109. De Bolle, X. et al. The length of a tetranucleotide repeat tract in Haemophilus influenzae determines the phase variation rate of a gene with homology to type III DNA methyltransferases. Mol. Microbiol. 35, 211–222 (2000).

    PubMed  Google Scholar 

  110. de Vries, N. et al. Transcriptional phase variation of a type III restriction-modification system in Helicobacter pylori. J. Bacteriol. 184, 6615–6623 (2002).

    PubMed  PubMed Central  Google Scholar 

  111. Srikhanta, Y. N., Maguire, T. L., Stacey, K. J., Grimmond, S. M. & Jennings, M. P. The phasevarion: a genetic system controlling coordinated, random switching of expression of multiple genes. Proc. Natl Acad. Sci. USA 102, 5547–5551 (2005).

    CAS  PubMed  Google Scholar 

  112. Fox, K. L. et al. Haemophilus influenzae phasevarions have evolved from type III DNA restriction systems into epigenetic regulators of gene expression. Nucleic Acids Res. 35, 5242–5252 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Srikhanta, Y. N. et al. Phasevarions mediate random switching of gene expression in pathogenic Neisseria. PLOS Pathog. 5, e1000400 (2009).

    PubMed  PubMed Central  Google Scholar 

  114. Srikhanta, Y. N., Fox, K. L. & Jennings, M. P. The phasevarion: phase variation of type III DNA methyltransferases controls coordinated switching in multiple genes. Nat. Rev. Microbiol. 8, 196–206 (2010).

    CAS  PubMed  Google Scholar 

  115. Kwiatek, A., Mrozek, A., Bacal, P., Piekarowicz, A. & Adamczyk-Poplawska, M. Type III methyltransferase M.NgoAX from Neisseria gonorrhoeae FA1090 regulates biofilm formation and interactions with human cells. Front. Microbiol. 6, 1426 (2015).

    PubMed  PubMed Central  Google Scholar 

  116. Blakeway, L. V. et al. Moraxella catarrhalis restriction-modification systems are associated with phylogenetic lineage and disease. Genome Biol. Evol. 10, 2932–2946 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Atack, J. M., Yang, Y., Seib, K. L., Zhou, Y. & Jennings, M. P. A survey of Type III restriction-modification systems reveals numerous, novel epigenetic regulators controlling phase-variable regulons: phasevarions. Nucleic Acids Res. 46, 3532–3542 (2018). A comprehensive bioinformatic search for phase-variable type III R-M DNA methyltransferases.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Gawthorne, J. A., Beatson, S. A., Srikhanta, Y. N., Fox, K. L. & Jennings, M. P. Origin of the diversity in DNA recognition domains in phasevarion associated modA genes of pathogenic Neisseria and Haemophilus influenzae. PLOS ONE 7, e32337 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Srikhanta, Y. N. et al. Methylomic and phenotypic analysis of the ModH5 phasevarion of Helicobacter pylori. Sci. Rep. 7, 16140 (2017).

    PubMed  PubMed Central  Google Scholar 

  120. Anjum, A. et al. Phase variation of a Type IIG restriction-modification enzyme alters site-specific methylation patterns and gene expression in Campylobacter jejuni strain NCTC11168. Nucleic Acids Res. 44, 4581–4594 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Sitaraman, R., Denison, A. M. & Dybvig, K. A unique, bifunctional site-specific DNA recombinase from Mycoplasma pulmonis. Mol. Microbiol. 46, 1033–1040 (2002).

    CAS  PubMed  Google Scholar 

  122. Doberenz, S. et al. Identification of a Pseudomonas aeruginosa PAO1 DNA methyltransferase, its targets, and physiological roles. MBio 8, e02312–e02316 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Manso, A. S. et al. A random six-phase switch regulates pneumococcal virulence via global epigenetic changes. Nat. Commun. 5, 5055 (2014). This study, together with Li et al. (2016) and Oliver et al. (2017), characterizes a phase variation mechanism that controls switching between pneumococcal phenotypic forms.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Li, J. et al. Epigenetic switch driven by DNA inversions dictates phase variation in Streptococcus pneumoniae. PLOS Pathog. 12, e1005762 (2016).

    PubMed  PubMed Central  Google Scholar 

  125. Oliver, M. B., Basu Roy, A., Kumar, R., Lefkowitz, E. J. & Swords, W. E. Streptococcus pneumoniae TIGR4 phase-locked opacity variants differ in virulence phenotypes. mSphere 2, e00386-17 (2017).

    PubMed  PubMed Central  Google Scholar 

  126. Estibariz, I. et al. The core genome m5C methyltransferase JHP1050 (M.Hpy99III) plays an important role in orchestrating gene expression in Helicobacter pylori. Nucleic Acids Res. 47, 2336–2348 (2019). This article demonstrates direct transcriptional control of bacterial genes by 5mC.

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Chao, M. C. et al. A cytosine methyltransferase modulates the cell envelope stress response in the cholera pathogen. PLOS Genet. 11, e1005666 (2015).

    PubMed  PubMed Central  Google Scholar 

  128. Kahramanoglou, C. et al. Genomics of DNA cytosine methylation in Escherichia coli reveals its role in stationary phase transcription. Nat. Commun. 3, 886 (2012).

    PubMed  Google Scholar 

  129. Militello, K. T., Mandarano, A. H., Varechtchouk, O. & Simon, R. D. Cytosine DNA methylation influences drug resistance in Escherichia coli through increased sugE expression. FEMS Microbiol. Lett. 350, 100–106 (2014).

    CAS  PubMed  Google Scholar 

  130. Kumar, S. et al. N4-cytosine DNA methylation regulates transcription and pathogenesis in Helicobacter pylori. Nucleic Acids Res. 46, 3429–3445 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Budroni, S. et al. Neisseria meningitidis is structured in clades associated with restriction modification systems that modulate homologous recombination. Proc. Natl Acad. Sci. USA 108, 4494–4499 (2011).

    CAS  PubMed  Google Scholar 

  132. Nandi, T. et al. Burkholderia pseudomallei sequencing identifies genomic clades with distinct recombination, accessory, and epigenetic profiles. Genome Res. 25, 608 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Kobayashi, I. Behavior of restriction-modification systems as selfish mobile elements and their impact on genome evolution. Nucleic Acids Res. 29, 3742–3756 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Tomcsanyi, T. & Berg, D. E. Transposition effect of adenine (Dam) methylation on activity of O end mutants of IS50. J. Mol. Biol. 209, 191–193 (1989).

    CAS  PubMed  Google Scholar 

  135. Alonso, A., Pucciarelli, M. G., Figueroa-Bossi, N. & Garcia-del Portillo, F. Increased excision of the Salmonella prophage ST64B caused by a deficiency in Dam methylase. J. Bacteriol. 187, 7901–7911 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Murphy, K. C., Ritchie, J. M., Waldor, M. K., Lobner-Olesen, A. & Marinus, M. G. Dam methyltransferase is required for stable lysogeny of the Shiga toxin (Stx2)-encoding bacteriophage 933W of enterohemorrhagic Escherichia coli O157:H7. J. Bacteriol. 190, 438–441 (2008).

    CAS  PubMed  Google Scholar 

  137. Wang, T. C. & Smith, K. C. Inviability of dam recA and dam recB cells of Escherichia coli is correlated with their inability to repair DNA double-strand breaks produced by mismatch repair. J. Bacteriol. 165, 1023–1025 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Veening, J. W., Smits, W. K. & Kuipers, O. P. Bistability, epigenetics, and bet-hedging in bacteria. Annu. Rev. Microbiol. 62, 193–210 (2008).

    CAS  PubMed  Google Scholar 

  139. Maynard-Smith, J. Evolution and the Theory of Games (Cambridge University Press, 1982).

  140. Roth, J. R., Kugelberg, E., Reams, A. B., Kofoid, E. & Andersson, D. I. Origin of mutations under selection: the adaptive mutation controversy. Annu. Rev. Microbiol. 60, 477–501 (2006).

    CAS  PubMed  Google Scholar 

  141. Tanner, J. R. & Kingsley, R. A. Evolution of Salmonella within hosts. Trends Microbiol. 26, 986–998 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Turrientes, M. C. et al. Normal mutation rate variants arise in a mutator (MutS) Escherichia coli population. PLOS ONE 8, e72963 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Furuta, Y. & Kobayashi, I. Movement of DNA sequence recognition domains between non-orthologous proteins. Nucleic Acids Res. 40, 9218–9232 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Coulondre, C., Miller, J. H., Farabaugh, P. J. & Gilbert, W. Molecular basis of base substitution hotspots in Escherichia coli. Nature 274, 775–780 (1978).

    CAS  PubMed  Google Scholar 

  145. Lieb, M. & Bhagwat, A. S. Very short patch repair: reducing the cost of cytosine methylation. Mol. Microbiol. 20, 467–473 (1996).

    CAS  PubMed  Google Scholar 

  146. Hershberg, R. & Petrov, D. A. Evidence that mutation is universally biased towards AT in bacteria. PLOS Genet. 6, e1001115 (2011).

    Google Scholar 

  147. Lobry, J. R. & Sueoka, N. Asymmetric directional mutation pressures in bacteria. Genome Biol. 3, RESEARCH0058 (2002).

    PubMed  PubMed Central  Google Scholar 

  148. Pereira, J. M., Hamon, M. A. & Cossart, P. A lasting impression: epigenetic memory of nacterial infections? Cell Host Microbe. 19, 579–582 (2016).

    CAS  PubMed  Google Scholar 

  149. Niller, H. H. & Minarovits, J. Patho-epigenetics of infectious diseases caused by intracellular bacteria. Adv. Exp. Med. Biol. 879, 107–130 (2016).

    CAS  PubMed  Google Scholar 

  150. Bierne, H. in Epigenetics of Infectious Diseases (eds Doerfler, W. & Casadesus, J.) 113–158 (Springer, 2017).

  151. Chernov, A. V. et al. Mycoplasma CG- and GATC-specific DNA methyltransferases selectively and efficiently methylate the host genome and alter the epigenetic landscape in human cells. Epigenetics 10, 303–318 (2015).

    PubMed  PubMed Central  Google Scholar 

  152. Mashhoon, N., Pruss, C., Carroll, M., Johnson, P. H. & Reich, N. O. Selective inhibitors of bacterial DNA adenine methyltransferases. J. Biomol. Screen 11, 497–510 (2006).

    CAS  PubMed  Google Scholar 

  153. Benkovic, S. J. et al. Identification of borinic esters as inhibitors of bacterial cell growth and bacterial methyltransferases, CcrM and MenH. J. Med. Chem. 48, 7468–7476 (2005).

    CAS  PubMed  Google Scholar 

  154. Ceccaldi, A. et al. Identification of novel inhibitors of DNA methylation by screening of a chemical library. ACS Chem. Biol. 8, 543–548 (2013).

    CAS  PubMed  Google Scholar 

  155. Cohen, N. R. et al. A role for the bacterial GATC methylome in antibiotic stress survival. Nat. Genet. 48, 581–586 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Maier, J. A. H., Mohrle, R. & Jeltsch, A. Design of synthetic epigenetic circuits featuring memory effects and reversible switching based on DNA methylation. Nat. Commun. 8, 15336 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Olivenza, D. R. et al. A portable epigenetic switch for bistable gene expression in bacteria. Sci. Rep. 9, 11261 (2019).

    PubMed  PubMed Central  Google Scholar 

  158. Nou, X. et al. Regulation of pyelonephritis-associated pili phase-variation in Escherichia coli: binding of the PapI and the Lrp regulatory proteins is controlled by DNA methylation. Mol. Microbiol. 7, 545–553 (1993).

    CAS  PubMed  Google Scholar 

  159. Heithoff, D. M. et al. Salmonella DNA adenine methylase mutants confer cross-protective immunity. Infect. Immun. 69, 6725–6730 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Heithoff, D. M., House, J. K., Thomson, P. C. & Mahan, M. J. Development of a Salmonella cross-protective vaccine for food animal production systems. Vaccine 33, 100–107 (2015).

    CAS  PubMed  Google Scholar 

  161. Xiao, C. L. et al. N6-methyladenine DNA modification in the human genome. Mol. Cell 71, 306–318.e7 (2018).

    CAS  PubMed  Google Scholar 

  162. Lopez-Garrido, J. & Casadesus, J. Crosstalk between virulence loci: regulation of Salmonella enterica pathogenicity island 1 (SPI-1) by products of the std fimbrial operon. PLOS ONE 7, e30499 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Braaten, B. A. et al. Leucine-responsive regulatory protein controls the expression of both the pap and fan pili operons in Escherichia coli. Proc. Natl Acad. Sci. USA 89, 4250–4254 (1992).

    CAS  PubMed  Google Scholar 

  164. Waldminghaus, T., Weigel, C. & Skarstad, K. Replication fork movement and methylation govern SeqA binding to the Escherichia coli chromosome. Nucleic Acids Res. 40, 5465–5476 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Badie, G., Heithoff, D. M. & Mahan, M. J. LcrV synthesis is altered by DNA adenine methylase overproduction in Yersinia pseudotuberculosis and is required to confer immunity in vaccinated hosts. Infect. Immun. 72, 6707–6710 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Pucciarelli, M. G., Prieto, A. I., Casadesus, J. & Garcia-del Portillo, F. Envelope instability in DNA adenine methylase mutants of Salmonella enterica. Microbiology 148, 1171–1182 (2002).

    CAS  PubMed  Google Scholar 

  167. Fumeaux, C. et al. Cell cycle transition from S-phase to G1 in Caulobacter is mediated by ancestral virulence regulators. Nat. Commun. 5, 4081 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Work in the authors’ laboratory is supported by grant BIO2016-75235-P from the Ministerio de Ciencia, Innovación y Universidades of Spain and the European Regional Fund. They are grateful to M. van der Woude and L. García-Pastor for discussions.

Author information

Authors and Affiliations

Authors

Contributions

J.C. and M.A.S.-R. contributed to discussion of the content, wrote the article, reviewed and edited the manuscript before submission, and researched data for the article.

Corresponding author

Correspondence to Josep Casadesús.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Microbiology thanks M. R. Oggioni and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Methylome

Overall DNA methylation pattern in a genome.

Adaptive value

In population genetics, the contribution of a phenotypic trait to the fitness of an individual or a population.

Restriction–modification (R-M) systems

Machine systems that eliminate foreign DNA molecules by endonucleolytic cleavage and protect the genome by modification of the cognate endonuclease target. A frequent type of protection is DNA methylation.

SOS regulon

Regulatory network that responds to DNA damage in gammaproteobacteria.

Homopolymeric nucleotide tract

DNA region that contains only AT or GC nucleotide pairs.

Transparent–opaque transition

Formation of phenotypic variants (phase variation) of Streptococcus pneumoniae that is involved in pneumococcal carriage and invasive infection.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sánchez-Romero, M.A., Casadesús, J. The bacterial epigenome. Nat Rev Microbiol 18, 7–20 (2020). https://doi.org/10.1038/s41579-019-0286-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-019-0286-2

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